1
|
Ma S, Su S, Zhang X, Wang X, Yi H. CircRNA encoded-peptide: Potential stock in the transcriptomics market. Life Sci 2025; 372:123643. [PMID: 40246192 DOI: 10.1016/j.lfs.2025.123643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/03/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
The emergence of circRNA-encoded peptides has sparked significant debate in recent years as a novel mode of action for circRNAs. A mounting body of evidence suggests that these peptides play vital roles in cancer development and immune responses. This review initially elucidates the presence of circRNA-encoded peptides and delineates their specific functions across various biological processes and pathological conditions. It goes on to furnish illustrative instances to underscore the pivotal involvement of circRNA-encoded peptides in both innate and adaptive immune responses. The study sheds new light on the biological roles of circRNAs, their potential tumor-promoting and tumor-suppressing functions of circRNA-encoded peptides in specific tumor environment, and their significance in immunological contexts. Meanwhile, the limitations of existing studies on circRNA-encoded peptides are discussed in depth. In particular, circRNA-encoded peptides are critically analyzed as biomarkers and therapeutic targets. Intriguingly, the review concludes with a more organized discussion of future research on circRNA-encoded peptides.
Collapse
Affiliation(s)
- Siyuan Ma
- Central Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin 130031, China; Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin 130021, China
| | - Sensen Su
- Central Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin 130031, China; Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin 130021, China; Department of Nephrology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xiuna Zhang
- Central Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin 130031, China; Department of Gastroenterology, Lequn Branch, The First Hospital of Jilin University, Changchun 130000, China
| | - Xiangxiu Wang
- Department of Otorhinolaryngology-Head and Neck Surgery, Gongli Hospital of Pudong New Area, Shanghai 200135, China
| | - Huanfa Yi
- Central Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin 130031, China; Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin 130021, China.
| |
Collapse
|
2
|
Li J, Xu S, Zhan Y, Lv X, Sun Z, Man L, Yang D, Sun Y, Ding S. CircRUNX1 enhances the Warburg effect and immune evasion in non-small cell lung cancer through the miR-145/HK2 pathway. Cancer Lett 2025; 620:217639. [PMID: 40090573 DOI: 10.1016/j.canlet.2025.217639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/18/2025]
Abstract
Non-small cell lung cancer (NSCLC) is acknowledged as the primary subtype of lung cancer. The Warburg effect, marked by elevated glucose consumption and lactate fermentation, is a prevalent characteristic of NSCLC. The mechanisms by which circRNA mediates the regulation of the Warburg effect and immune evasion in NSCLC remain unclear. This study found an elevated circRNA, circRUNX1, whiche promotes glycolysis and lactate generation, resulting in the infiltration of regulatory T cell (Treg) in NSCLC. circRUNX1 acts as a miR-145 sponge, inhibiting its negative regulation of the target gene HK2, therefore facilitating glycolysis and lactate generation. The accumulation of lactic acid in the tumor microenvironment promotes Treg cell proliferation and aids immune evasion. Functionally, the suppression of circRUNX1 significantly impedes tumor development both in vitro and in vivo. These findings collectively clarity a previously unexamined mechanism linking the circRUNX1/miR-145/HK2 axis in regulation of the Warburg effect and immune evasion in NSCLC.
Collapse
MESH Headings
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Humans
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Warburg Effect, Oncologic
- Animals
- Tumor Microenvironment/immunology
- Mice
- Hexokinase/genetics
- Hexokinase/metabolism
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
- Immune Evasion
- Cell Proliferation
- Glycolysis
- T-Lymphocytes, Regulatory/immunology
- Signal Transduction
- Lactic Acid/metabolism
- Tumor Escape
- Female
Collapse
Affiliation(s)
- Jinyou Li
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China; Department of Thoracic Surgery, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Shiwei Xu
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China; Department of Thoracic Surgery, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Yangyang Zhan
- Department of Pharmacy, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, 225 Changhai Road, Yangpu District, Shanghai, China; Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Xinyi Lv
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, China; School of Medicine, Nantong University, Nantong, 226001, China
| | - Zhenyu Sun
- Department of Thoracic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, 214000, China; Department of Thoracic Surgery, Wuxi School of Medicine, Jiangnan University, Wuxi, 214000, China
| | - Li Man
- Department of Medical Oncology, Anshan Cancer Hospital, Anshan, 114000, China
| | - Donghua Yang
- New York College of Traditional Chinese Medicine, 200 Old Country Rd, Suite 500, Mineola, NY, 11501, USA
| | - Yahong Sun
- Department of Respiratory and Critical Care Medicine, Haining People's Hospital, Haining, 314400, China.
| | - Shengguang Ding
- Department of Thoracic Surgery, Affiliated Hospital 2 of Nantong University, Nantong First People's Hospital, Nantong, 226001, China.
| |
Collapse
|
3
|
Liu Y, Si L, Liu Y, Li S, Zhang X, Jiang S, Liu W, Li X, Zhang L, Zheng H, Liu Z, Hu J, Chen J. Construction of a programmed activation nanosystem based on intracellular hypoxia in cisplatin-resistant tumor cells for reversing cisplatin resistance. Mater Today Bio 2025; 32:101709. [PMID: 40230650 PMCID: PMC11995088 DOI: 10.1016/j.mtbio.2025.101709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer poses a significant threat to human life and health. Cancers treated with cisplatin invariably develop drug resistance. This challenge can be overcome by identifying and exploiting the vulnerabilities acquired by drug-resistant cancer cells, paving the way for finding effective novel treatment options for cisplatin-resistant cancers. Our previous study revealed that cisplatin resistance in cancer cells comes at the cost of increased intracellular hypoxia. In this study, we used 2-nitroimidazole modified hyaluronic acid (HA-NI) as the carrier. The cisplatin-resistant tumor cell specific intracellular hypoxia programmed activation nanomedicine (T/C@HN NPs) was constructed by the hypoxic toxic drug tirapazamine (TPZ) and encapsulating chlorin e6 (Ce6) into HA-NI using polymer assembly technology. The amphiphilic carrier could release free Ce6 molecules under the stimulation of intracellular hypoxic environment, and exhibit specific "activated state" photodynamic properties in cisplatin-resistant tumor cells. Upon irradiation, Ce6-mediated photodynamic therapy further intensifies hypoxia, amplifying its cytotoxicity. This project systematically evaluated the effects of T/C@HN NPs on the identification and recognition of cisplatin-resistant tumors using drug-resistant patient-derived xenograft (PDX) models. This study provides a promising avenue for the development of novel treatment of cisplatin-resistant tumors.
Collapse
Affiliation(s)
- Yurong Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Longqing Si
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Yunheng Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Song Li
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xiaokang Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Shaojing Jiang
- Yantai Engineering Research Center for Digital Technology of Stomatology, School of Stomatology, Binzhou Medical University, Yantai, 264003, China
| | - Wenjing Liu
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Xiaolin Li
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Lianguo Zhang
- Department of Thoracic Surgery, Binzhou Medical University Hospital, Binzhou, 256603, China
| | - Hongxia Zheng
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| | - Zhonghao Liu
- Yantai Engineering Research Center for Digital Technology of Stomatology, School of Stomatology, Binzhou Medical University, Yantai, 264003, China
| | - Jinghui Hu
- Yantai Engineering Research Center for Digital Technology of Stomatology, School of Stomatology, Binzhou Medical University, Yantai, 264003, China
| | - Jing Chen
- School of Pharmacy, Binzhou Medical University, Yantai, 264003, China
| |
Collapse
|
4
|
Liu G, Liu J, Li S, Zhang Y, He R. Exosome-Mediated Chemoresistance in Cancers: Mechanisms, Therapeutic Implications, and Future Directions. Biomolecules 2025; 15:685. [PMID: 40427578 PMCID: PMC12108986 DOI: 10.3390/biom15050685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Chemotherapy resistance represents a formidable obstacle in oncological therapeutics, substantially compromising the efficacy of adjuvant chemotherapy regimens and contributing to unfavorable clinical prognoses. Emerging evidence has elucidated the pivotal involvement of exosomes in the dissemination of chemoresistance phenotypes among tumor cells and within the tumor microenvironment. This review delineates two distinct intra-tumoral resistance mechanisms orchestrated by exosomes: (1) the exosome-mediated sequestration of chemotherapeutic agents coupled with enhanced drug efflux in neoplastic cells, and (2) the horizontal transfer of chemoresistance to drug-sensitive cells through the delivery of bioactive molecular cargo, thereby facilitating the propagation of resistance phenotypes across the tumor population. Furthermore, the review covers current in vivo experimental data focusing on targeted interventions against specific genetic elements and exosomal secretion pathways, demonstrating their potential in mitigating chemotherapy resistance. Additionally, the therapeutic potential of inhibiting exosome-mediated transporter transfer strategy is particularly examined as a promising strategy to overcome tumor resistance mechanisms.
Collapse
Affiliation(s)
| | | | | | - Yumiao Zhang
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| | - Ren He
- School of Chemical Engineering and Technology, School of Synthetic Biology and Biomanufacturing, Frontiers Science Center for Synthetic Biology (Ministry of Education) and State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin 300350, China; (G.L.); (J.L.); (S.L.)
| |
Collapse
|
5
|
Sristi, Gupta G, Abourehab MAS, Sahebkar A, Kesharwani P. Recent advances in PD-L1 siRNA nanocarriers for cancer therapy. Int J Biol Macromol 2025; 311:143994. [PMID: 40339860 DOI: 10.1016/j.ijbiomac.2025.143994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Tumor immune evasion depends on the programmed death-ligand 1 (PD-L1) mechanism, making it a prominent target in cancer therapy. Small interfering RNA (siRNA) designed to inhibit PD-L1 expression presents an innovative approach for boosting immunity against tumors. However, the therapeutic use of siRNA faces challenges, primarily due to its instability and inefficient cellular delivery. Recent advancements in nanocarrier technologies have shown promise in overcoming these obstacles, improving the delivery and efficacy of PD-L1 siRNA. This review comprehensively explores various nanocarrier systems, including lipid nanoparticles, polymeric carriers, and inorganic nanoparticles, highlighting their design innovations and applications in targeting PD-L1 in diverse cancer models. We discuss the synergistic effects of PD-L1 siRNA delivered via nanocarriers in conjunction with chemotherapy and immunomodulators, showcasing their potential to boost immune responses and reduce tumor growth. Additionally, we address ongoing challenges such as optimizing biodistribution and minimizing off-target effects, which hinder clinical translation. By synthesizing recent research findings, this review aims to illuminate the transformative potential of PD-L1 siRNA nanocarriers in cancer immunotherapy, paving the way for future studies aimed at enhancing therapeutic strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Sristi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Sector 125, Noida 201301, UP, India
| | - Garima Gupta
- Graphic Era Hill University, Dehradun 248002, India; School of Allied Medical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Mohammed A S Abourehab
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| |
Collapse
|
6
|
Mai Z, Chen X, Lu Y, Zheng J, Lin Y, Lin P, Zheng Y, Zhou Z, Xu R, Guo B, Cui L, Zhao X. Orchestration of immunoregulatory signaling ligand and receptor dynamics by mRNA modifications: Implications for therapeutic potential. Int J Biol Macromol 2025; 310:142987. [PMID: 40210040 DOI: 10.1016/j.ijbiomac.2025.142987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/26/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
RNA modifications are pivotal regulators of gene expression, significantly influencing immune responses by modulating the stability and translation of mRNAs encoding key immunoregulatory ligands and receptors. Among these modifications, N6-methyladenosine (m6A) is the most abundant and well-characterized, orchestrating immune evasion, T-cell exhaustion, and cytokine production by dynamically regulating transcripts such as PD-L1, IFN-γ, and TGF-β. These modifications critically impact the function and availability of proteins essential for maintaining immune homeostasis and shaping adaptive immune responses. This review comprehensively examines established and emerging roles of mRNA modifications in regulating immunoregulatory signaling, including co-inhibitory and co-stimulatory molecules, chemokines, cytokines, and transforming growth factor-β. We highlight how m6A writers, erasers, and readers finely regulate immune checkpoints and inflammatory pathways across cancer, infection, and autoimmune diseases. Furthermore, the review provides a critical analysis of current discrepancies in the field, emphasizing factors contributing to inconsistencies and offering insights into the complex nature of epigenetic regulation. Challenges and limitations in this rapidly evolving area are also discussed. Advancing detection technologies and developing specific inhibitors targeting RNA-modifying proteins will be crucial for precisely modulating immune responses, paving the way for innovations in precision medicine and immunotherapy.
Collapse
Affiliation(s)
- Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Xu Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Yucheng Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Zihao Zhou
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Rongwei Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China
| | - Bing Guo
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, Guangdong, China
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China; School of Dentistry, University of California, Los Angeles, Los Angeles 90095, CA, USA.
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou 510280, Guangdong, China.
| |
Collapse
|
7
|
Saadh MJ, Ghnim ZS, Mahdi MS, Mandaliya V, Ballal S, Bareja L, Chaudhary K, Sharma R, Gupta S, Taher WM, Alwan M, Jawad MJ, Hamad AK. The emerging role of kinesin superfamily proteins in Wnt/β-catenin signaling: Implications for cancer. Pathol Res Pract 2025; 269:155904. [PMID: 40073645 DOI: 10.1016/j.prp.2025.155904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Cellular processes such as proliferation, differentiation, and tissue homeostasis are significantly influenced by the Wnt/β-catenin signaling pathway. Dysregulation of this pathway has been implicated in the development of various types of cancer. This study focuses on the emerging role of kinesin superfamily proteins (KIFs) in modulating cancer signaling. KIFs, a group of motor proteins, have attracted attention for their dual roles in intracellular transport: facilitating the cellular entry of Wnt ligands and contributing to the assembly of the β-catenin destruction complex. The study explores the interactions between KIFs and the Wnt/β-catenin pathway, identifying specific KIFs that interact with key components of the signaling cascade and examining their roles in cancer progression. Furthermore, it evaluates therapeutic strategies targeting KIFs to suppress aberrant Wnt activity in cancer and investigates how KIF-mediated transport spatially and temporally regulates Wnt signaling. The insights provided could guide future research into the role of KIFs in cancer biology and their involvement in oncogenic signaling pathways.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Viralkumar Mandaliya
- Marwadi University Research Center, Department of Microbiology, Faculty of Science Marwadi University, Rajkot, Gujarat 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Rsk Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Sofia Gupta
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
8
|
Das A, Sonar S, Dhar R, Subramaniyan V. Exosomes in melanoma: Future potential for clinical theranostics. Pathol Res Pract 2025; 269:155950. [PMID: 40179441 DOI: 10.1016/j.prp.2025.155950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/13/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Melanoma, an aggressive form of skin cancer, presents significant therapeutic challenges due to its resistance to conventional treatments and propensity for metastasis. Exosomes, nanoscale vesicles secreted by a wide variety of cells, have emerged as promising tools for developing novel melanoma therapies. Exosome-based therapeutic approaches offer several advantages, including inherent biocompatibility, low immunogenicity, and the ability to cross biological barriers. This review explores the therapeutic potential of exosomes in melanoma treatment, focusing on their multifaceted roles in modulating tumor cell behavior, enhancing anti-tumor immune responses, and serving as targeted drug delivery vehicles. We discuss various strategies employed to engineer exosomes for enhanced therapeutic efficacy, including loading them with chemotherapeutic agents, small interfering RNAs (siRNAs), microRNAs (miRNAs), and immunomodulatory molecules. Additionally, we highlight the potential of exosomes derived from diverse sources to enhance anti-cancer effects. Furthermore, we address the challenges and future directions in translating exosome-based therapies from bench to bedside, emphasizing the need for standardized isolation and manufacturing protocols, as well as rigorous preclinical and clinical evaluations to unlock the full therapeutic potential of exosomes in the fight against melanoma.
Collapse
Affiliation(s)
- Asmit Das
- Department of Oncology and Maxillofacial Pathology, Neuron Institute of Applied Research, Amravati, Maharashtra, India
| | - Swarup Sonar
- Department of Oncology and Maxillofacial Pathology, Neuron Institute of Applied Research, Amravati, Maharashtra, India
| | - Rajib Dhar
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya, Selangor 47500, Malaysia
| | - Vetriselvan Subramaniyan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya, Selangor 47500, Malaysia.
| |
Collapse
|
9
|
Wen Z, Zhang W, Wu W. The latest applications of exosome-mediated drug delivery in anticancer therapies. Colloids Surf B Biointerfaces 2025; 249:114500. [PMID: 39799609 DOI: 10.1016/j.colsurfb.2025.114500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 01/15/2025]
Abstract
In recent years, the significant role of anticancer drugs in cancer treatment has garnered considerable attention. However, the application of these drugs is largely limited by their short half-life in blood circulation, low cellular uptake efficiency, and off-target effects. Exosomes, which serve as crucial messengers in intercellular communication, exhibit unique advantages in molecular delivery compared to traditional synthetic carriers, thereby offering new possibilities for modern drug delivery systems. Exosomes possess organotropic functions and are naturally produced by cells, making them promising candidates for natural drug delivery systems with organotropic properties and minimal side effects. These naturally derived carriers can achieve stable, efficient, and selective delivery of anticancer drugs, thereby enhancing the efficacy and potential of anticancer agents in cancer immunotherapy. This review provides a concise overview of the unique characteristics of exosomes related to anticancer drug delivery, strategies for utilizing exosomes as carriers in cancer therapy, and the latest advancements in the field.
Collapse
Affiliation(s)
- Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
10
|
Yin H, Shi J, Li S, You Q, Zhu H, Koo C, Liu B, Hou L, Wu C. Emerging roles of exosomal circRNAs in non-small cell lung cancer. J Transl Med 2025; 23:490. [PMID: 40307927 PMCID: PMC12042431 DOI: 10.1186/s12967-025-06463-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/06/2025] [Indexed: 05/02/2025] Open
Abstract
Despite the prevalence of non-small cell lung cancer (NSCLC) is high, the limited early detection and management of these tumors are restricted since there is an absence of reliable and precise diagnostic biomarkers and therapeutic targets. Exosomes transport functional molecules for facilitating intercellular communication, especially in the tumor microenvironment, indicating their potential as cancer biomarkers and therapeutic targets. Circular RNA (circRNA), a type of non-coding RNA possessing a covalently closed loop structure, substantial abundance, and tissue-specific expression patterns, is stably enriched in exosomes. In recent years, significant breakthroughs have been made in research on exosomal circRNA in NSCLC. This review briefly introduces the biogenesis, characterizations, and functions of circRNAs and exosomes, and systematically describes the biological functions and mechanisms of exosomal circRNAs in NSCLC. In addition, this study summarizes their role in the progression of NSCLC and discusses their clinical significance as biomarkers and therapeutic targets for NSCLC.
Collapse
Affiliation(s)
- Hongyuan Yin
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jiayi Shi
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shaoling Li
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qianhui You
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huici Zhu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chinying Koo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Baonian Liu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
11
|
Khalil EIM, Mohamed FEZA, Kamal Mohamed R. Upregulated ATG4B predicts poor prognosis and correlates with angiogenesis in osteosarcoma. J Egypt Natl Canc Inst 2025; 37:24. [PMID: 40279002 DOI: 10.1186/s43046-025-00269-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 03/04/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary bone cancer in children and adolescents. Between 35 and 45% of these patients do not respond to standard chemotherapeutic treatments, resulting in a very low 5-year survival rate of only 5-20%. This resistance often leads to treatment failure and unfavorable prognoses, highlighting the critical need for new therapeutic targets to improve treatment strategies. Autophagy-related gene 4 B (ATG4B) is a crucial cysteine protease for autophagosome formation. It is overexpressed and correlates with poor prognosis in various cancers. However, the relationship between ATG4B expression and angiogenesis in OS remains unexplored. This study investigated the expression levels of ATG4B and VEGF in OS and their correlation with clinicopathological data. MATERIALS AND METHODS This study included 67 paraffin-embedded OS tissue samples. ATG4B and VEGF expression levels were assessed via immunohistochemistry, and their associations with clinicopathological variables were statistically analyzed. Additionally, ATG4B gene expression in OS was examined via GEO datasets from https://www.ncbi.nlm.nih.gov . RESULTS ATG4B and VEGF were expressed in 79.1% and 74.6% of the osteosarcoma samples, respectively. There was a significant positive correlation between ATG4B expression and tumor size, tumor stage, and histological response to neoadjuvant chemotherapy, with p values of 0.013, 0.008, and 0.022, respectively. VEGF expression was also significantly correlated with tumor size, tumor stage, and the presence of distant metastasis at diagnosis, with p values of 0.022, 0.044, and 0.013, respectively. A notable positive correlation between ATG4B and VEGF expression levels was observed (p=0.002), which was supported by the GEO dataset analysis. High ATG4B and VEGF overexpression were significantly associated with worse overall survival by univariate analysis. CONCLUSIONS The results suggest that ATG4B acts as a tumor promoter in OS, indicating its potential as a therapeutic target to inhibit tumor growth. Elevated ATG4B levels may also serve as a marker for poor prognosis. Additionally, VEGF overexpression is linked to a greater likelihood of pulmonary metastasis and a worse overall prognosis. The positive correlation between ATG4B and VEGF suggests that the absence of both markers could be indicative of a better chemotherapy response, offering insights into potential new treatment approaches.
Collapse
Affiliation(s)
| | - Fatma El Zahraa Ammar Mohamed
- Pathological Sciences Department, MBBS Program,, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
- Pathology Department, Faculty of Medicine, Minia University, Minia, Egypt
| | | |
Collapse
|
12
|
Wang C, Han X, Kong S, Zhang S, Ning H, Wu F. Deciphering the mechanisms of PARP inhibitor resistance in prostate cancer: Implications for precision medicine. Biomed Pharmacother 2025; 185:117955. [PMID: 40086424 DOI: 10.1016/j.biopha.2025.117955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
Prostate cancer is a leading malignancy among men. While early-stage prostate cancer can be effectively managed, metastatic prostate cancer remains incurable, with a median survival of 3-5 years. The primary treatment for advanced prostate cancer is androgen deprivation therapy (ADT), but resistance to ADT often leads to castrationresistant prostate cancer (CRPC), presenting a significant therapeutic challenge. The advent of precision medicine has introduced promising new treatments, including PARP inhibitors (PARPi), which target defects in DNA repair mechanisms in cancer cells. PARPi have shown efficacy in treating advanced prostate cancer, especially in patients with metastatic CRPC (mCRPC) harboring homologous recombination (HR)-associated gene mutations. Despite these advancements, resistance to PARPi remains a critical issue. Here, we explored the primary mechanisms of PARPi resistance in prostate cancer. Key resistance mechanisms include homologous recombination recovery through reverse mutations in BRCA genes, BRCA promoter demethylation, and non-degradation of mutated BRCA proteins. The tumor microenvironment and overactivation of the base excision repair pathway also play significant roles in bypassing PARPi-induced synthetic lethality. In addition, we explored the clinical implications and therapeutic strategies to overcome resistance,emphasizing the need for precision medicine approaches. Our findings highlight the need for comprehensive strategies to improve PARPi sensitivity and effectiveness,ultimately aiming to extend patient survival and improve the quality of life for those with advanced prostate cancer. As our understanding of PARPi resistance evolves, more diverse and effective individualized treatment regimens will emerge.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China
| | - Xiaoran Han
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Shaoqiu Kong
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Shanhua Zhang
- Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China
| | - Hao Ning
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China; Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China.
| | - Fei Wu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, PR China; Department of Urology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, PR China.
| |
Collapse
|
13
|
Tang Z, Chen C, Zhou C, Liu Z, Li T, Zhang Y, Feng Y, Gu C, Li S, Chen J. Insights into tumor-derived exosome inhibition in cancer therapy. Eur J Med Chem 2025; 285:117278. [PMID: 39823808 DOI: 10.1016/j.ejmech.2025.117278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/11/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Exosomes are critical mediators of cell-to-cell communication in physiological and pathological processes, due to their ability to deliver a variety of bioactive molecules. Tumor-derived exosomes (TDEs), in particular, carry carcinogenic molecules that contribute to tumor progression, metastasis, immune escape, and drug resistance. Thus, TDE inhibition has emerged as a promising strategy to combat cancer. In this review, we discuss the key mechanisms of TDE biogenesis and secretion, emphasizing their implications in tumorigenesis and cancer progression. Moreover, we provide an overview of small-molecule TDE inhibitors that target specific biogenesis and/or secretion pathways, highlighting their potential use in cancer treatment. Lastly, we present the existing obstacles and propose corresponding remedies for the future development of TDE inhibitors.
Collapse
Affiliation(s)
- Ziwei Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States
| | - Zhouyan Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Zhang
- School of Petrochemical Engineering, Changzhou University, Changzhou, 213164, China.
| | - Yanyan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chenglei Gu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shijia Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jichao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
14
|
Zhao L, Gui Y, Deng X. Focus on mechano-immunology: new direction in cancer treatment. Int J Surg 2025; 111:2590-2602. [PMID: 39764598 DOI: 10.1097/js9.0000000000002224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/29/2024] [Indexed: 03/16/2025]
Abstract
The immune response is modulated by a diverse array of signals within the tissue microenvironment, encompassing biochemical factors, mechanical forces, and pressures from adjacent tissues. Furthermore, the extracellular matrix and its constituents significantly influence the function of immune cells. In the case of carcinogenesis, changes in the biophysical properties of tissues can impact the mechanical signals received by immune cells, and these signals c1an be translated into biochemical signals through mechano-transduction pathways. These mechano-transduction pathways have a profound impact on cellular functions, influencing processes such as cell activation, metabolism, proliferation, and migration, etc. Tissue mechanics may undergo temporal changes during the process of carcinogenesis, offering the potential for novel dynamic levels of immune regulation. Here, we review advances in mechanoimmunology in malignancy studies, focusing on how mechanosignals modulate the behaviors of immune cells at the tissue level, thereby triggering an immune response that ultimately influences the development and progression of malignant tumors. Additionally, we have also focused on the development of mechano-immunoengineering systems, with the help of which could help to further understand the response of tumor cells or immune cells to alterations in the microenvironment and may provide new research directions for overcoming immunotherapeutic resistance of malignant tumors.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
| | - Yajun Gui
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
| | - Xiangying Deng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, Changsha, Human, China
- Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
15
|
Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Front Immunol 2025; 16:1525052. [PMID: 40078996 PMCID: PMC11897508 DOI: 10.3389/fimmu.2025.1525052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Extracellular vesicles (EVs), as cell-derived small vesicles, facilitate intercellular communication within the tumor microenvironment (TME) by transporting biomolecules. EVs from different sources have varied contents, demonstrating differentiated functions that can either promote or inhibit cancer progression. Thus, regulating the formation, secretion, and intake of EVs becomes a new strategy for cancer intervention. Advancements in EV isolation techniques have spurred interest in EV-based therapies, particularly for tumor immunotherapy. This review explores the multifaceted functions of EVs from various sources in tumor immunotherapy, highlighting their potential in cancer vaccines and adoptive cell therapy. Furthermore, we explore the potential of EVs as nanoparticle delivery systems in tumor immunotherapy. Finally, we discuss the current state of EVs in clinical settings and future directions, aiming to provide crucial information to advance the development and clinical application of EVs for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xia
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunhan Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongen Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Zhang G, Wu X, Fu H, Sun D. Circular RNA microarray expression profile and potential function of circDOCK1 in colorectal cancer. Front Genet 2025; 16:1443876. [PMID: 39967686 PMCID: PMC11832710 DOI: 10.3389/fgene.2025.1443876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 01/13/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction Endoscopic tissue biopsy combined with histopathology is the gold standard for the diagnosis of colorectal cancer (CRC); however, the invasive nature of this procedure hinders its acceptance by patients. Therefore, there exists a critical need to identify novel markers facilitating early CRC detection and prognosis. Circular RNAs (circRNAs) hold promise as novel clinical diagnostic markers. This study aimed to investigate the impact of circDOCK1 on CRC metastasis and prognosis as well as its underlying molecular mechanisms. Methods We explored circRNA expression profiles in four pairs of CRC tissues and adjacent non-carcinoma tissues via microarray analysis. After Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and circRNA-miRNA network analyses, circDOCK1 was chosen for further investigation. We evaluated its clinical relevance in 80 CRC tissue pairs and adjacent controls, correlating circDOCK1 expression with clinical characteristics. Follow-up data from patient telephone interviews were analyzed for survival outcomes. Transfection efficiency was confirmed via qRT-PCR in HCT116 and SW480 colon cells, and the effects of circDOCK1 on cell proliferation, migration, and invasion were assessed. Results Microarray data revealed 149 significantly differentially expressed circRNAs, including 71 upregulated and 78 downregulated circRNAs, in CRC tissues. CircDOCK1 exhibited elevated expression in patients with CRC and emerged as an independent prognostic factor. Kaplan-Meier curve analysis suggested that circDOCK1 expression is an unfavorable prognostic factor in patients with CRC. In vivo experiments revealed that circDOCK1 overexpression enhanced the proliferation, migration, and invasion of CRC cells, with consistent results upon circDOCK1 downregulation. Conclusion These data indicate that circDOCK1 may play a role in promoting the proliferation, migration, and invasion of CRC cells, suggesting its potential as a CRC biomarker.
Collapse
Affiliation(s)
| | | | | | - Daqing Sun
- Department of Pediatric Surgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
17
|
Li X, Liu H, Xing P, Li T, Fang Y, Chen S, Dong S. Exosomal circRNAs: Deciphering the novel drug resistance roles in cancer therapy. J Pharm Anal 2025; 15:101067. [PMID: 39957900 PMCID: PMC11830318 DOI: 10.1016/j.jpha.2024.101067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/13/2024] [Accepted: 08/03/2024] [Indexed: 02/18/2025] Open
Abstract
Exosomal circular RNA (circRNAs) are pivotal in cancer biology, and tumor pathophysiology. These stable, non-coding RNAs encapsulated in exosomes participated in cancer progression, tumor growth, metastasis, drug sensitivity and the tumor microenvironment (TME). Their presence in bodily fluids positions them as potential non-invasive biomarkers, revealing the molecular dynamics of cancers. Research in exosomal circRNAs is reshaping our understanding of neoplastic intercellular communication. Exploiting the natural properties of exosomes for targeted drug delivery and disrupting circRNA-mediated pro-tumorigenic signaling can develop new treatment modalities. Therefore, ongoing exploration of exosomal circRNAs in cancer research is poised to revolutionize clinical management of cancer. This emerging field offers hope for significant breakthroughs in cancer care. This review underscores the critical role of exosomal circRNAs in cancer biology and drug resistance, highlighting their potential as non-invasive biomarkers and therapeutic targets that could transform the clinical management of cancer.
Collapse
Affiliation(s)
- Xi Li
- Department of Vascular and Thyroid Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Hanzhe Liu
- Department of Critical Care Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China
| | - Peiyu Xing
- Department of Ophthalmology, China Medical University the Fourth People's Hospital of Shenyang, Shenyang, 110031, China
| | - Tian Li
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Fang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Shuang Chen
- Department of Cardiology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Siyuan Dong
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
18
|
Guo X, Piao H, Sui R. Exosomes in the Chemoresistance of Glioma: Key Point in Chemoresistance. J Cell Mol Med 2025; 29:e70401. [PMID: 39950738 PMCID: PMC11826829 DOI: 10.1111/jcmm.70401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/17/2025] Open
Abstract
Gliomas are the most ordinary primary virulent brain tumours and commonly used clinical treatments include tumour resection, radiation therapy and chemotherapy. Although significant progress has been made in recent years in progression-free survival (PFS) and overall survival (OS) for patients with high-grade gliomas, the prognosis for patients remains poor. Chemoresistance refers to the phenomenon of decreased sensitivity of tumour cells to drugs, resulting in reduced or ineffective drug efficacy, and is an important cause of failure of tumour chemotherapy. Exosomes, a type of extracellular vesicle, are secreted by cancer cells and various stromal cells in the tumour microenvironment (TME) and transfer their inclusions to cancer cells, increasing chemoresistance. Furthermore, depletion of exosomes reverses certain detrimental effects on tumour metabolism and restores sensitivity to chemotherapeutic agents. Here, we summarised the correlation between exosomes and resistance to chemotherapeutic agents in glioma patients, the mechanisms of action of exosomes involved in resistance and their clinical value. We aimed to afford new thoughts for research, clinical diagnosis and intervention in the mechanisms of chemoresistance in glioma patients.
Collapse
Affiliation(s)
- Xu Guo
- Department of NeurosurgeryCancer Hospital of Daflian University of Technology, Liaoning Cancer Hospital & InstituteShenyangLiaoningChina
| | - Haozhe Piao
- Department of NeurosurgeryCancer Hospital of Daflian University of Technology, Liaoning Cancer Hospital & InstituteShenyangLiaoningChina
| | - Rui Sui
- Department of NeurosurgeryCancer Hospital of Daflian University of Technology, Liaoning Cancer Hospital & InstituteShenyangLiaoningChina
| |
Collapse
|
19
|
Li C, Zhao X, Zhao J, Zhao J, An L, Wu G. BRAF regulates circPSD3/miR-526b/RAP2A axis to hinder papillary thyroid carcinoma progression. BMC Mol Cell Biol 2025; 26:6. [PMID: 39838328 PMCID: PMC11753155 DOI: 10.1186/s12860-024-00528-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 12/27/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is a common malignant tumor. BRAFV600E mutation has become a common molecular event in PTC pathogenesis. Circular RNA PSD3 (circPSD3) is known to be highly expressed in PTC. However, the bio-functional role of circPSD3 and its possible relationship with the BRAF in PTC is not clear. This study aims to probe the biofunction and molecular mechanism of circPSD3 in PTC pathogenesis. METHODS RT-qPCR was utilized to measure the expression of circPSD3 and BRAF in PTC tissues and cells. The CCK-8 and EdU assays were employed to assess cell viability and proliferation. Cell apoptosis was quantified using flow cytometry. The migratory and invasive capabilities of the cells were evaluated via wound healing and transwell assays. The interaction between RNAs was investigated using luciferase reporter assay. Additionally, xenograft tumor experiments were conducted to validate our findings in vivo. RESULTS Data showed that circPSD3 was highly expressed in PTC patients and cell lines. CircPSD3 was found to promote cell growth and migration and inhibit apoptosis in PTC cells. Results also revealed that circPSD3 upregulated RAP2A expression by specifically sponging miR-526b. Interestingly, inhibiting miR-526b reversed the tumorigenic properties of circPSD3 in PTC. Additionally, BRAF expression was low in PTC patients, and overexpression of BRAF hampered PTC development by downregulating circPSD3 and RAP2A while upregulating miR-526b expressions. CONCLUSIONS Our study reveals that circPSD3 is a key regulator promoting PTC progression via the circPSD3/miR-526b/RAP2A pathway. Furthermore, we found that overexpressing BRAF, which inhibits circPSD3, significantly hampers the progression of PTC.
Collapse
Affiliation(s)
- Chuang Li
- Department of Ultrasound, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450000, China
| | - Xiaojuan Zhao
- Department of Ultrasound, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450000, China
| | - Jingge Zhao
- Department of Clinical Scientific Research Service Center, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450000, China
| | - Jing Zhao
- Department of Ultrasound, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450000, China
| | - Lemei An
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450000, China
| | - Gang Wu
- Department of Ultrasound, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450000, China.
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, No. 7 Weiwu Road, Jinshui District, Zhengzhou, Henan, 450000, China.
| |
Collapse
|
20
|
Zhang N, Wang X, Li Y, Lu Y, Sheng C, Sun Y, Ma N, Jiao Y. Mechanisms and therapeutic implications of gene expression regulation by circRNA-protein interactions in cancer. Commun Biol 2025; 8:77. [PMID: 39825074 PMCID: PMC11748638 DOI: 10.1038/s42003-024-07383-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 12/09/2024] [Indexed: 01/20/2025] Open
Abstract
Circular RNAs (circRNAs) have garnered substantial attention due to their distinctive circular structure and gene regulatory functions, establishing them as a significant class of functional non-coding RNAs in eukaryotes. Studies have demonstrated that circRNAs can interact with RNA-binding proteins (RBPs), which play crucial roles in tumorigenesis, metastasis, and drug response in cancer by influencing gene expression and altering the processes of tumor initiation and progression. This review aims to summarize the recent advances in research on circRNA-protein interactions (CPIs) and discuss the functions and mode of action of CPIs at various stages of gene expression, including transcription, splicing, translation, and post-translational modifications in the context of cancer. Additionally, we explore the role of CPIs in tumor drug resistance to gain a deeper understanding of their potential applications in the development of new anti-cancer therapeutic approaches.
Collapse
Affiliation(s)
- Nan Zhang
- Shengjing Hospital of China Medical University, Obstetrics and Gynecology Department, NO36. Sanhao Street, Heping district, Shenyang, China
| | - Xinjia Wang
- Shengjing Hospital of China Medical University, Obstetrics and Gynecology Department, NO36. Sanhao Street, Heping district, Shenyang, China
| | - Yu Li
- Shengjing Hospital of China Medical University, Obstetrics and Gynecology Department, NO36. Sanhao Street, Heping district, Shenyang, China
| | - Yiwei Lu
- Shengjing Hospital of China Medical University, Obstetrics and Gynecology Department, NO36. Sanhao Street, Heping district, Shenyang, China
| | - Chengcheng Sheng
- Shengjing Hospital of China Medical University, Obstetrics and Gynecology Department, NO36. Sanhao Street, Heping district, Shenyang, China
| | - Yumeng Sun
- Shengjing Hospital of China Medical University, Obstetrics and Gynecology Department, NO36. Sanhao Street, Heping district, Shenyang, China
| | - Ningye Ma
- Shengjing Hospital of China Medical University, Obstetrics and Gynecology Department, NO36. Sanhao Street, Heping district, Shenyang, China.
| | - Yisheng Jiao
- Shengjing Hospital of China Medical University, Obstetrics and Gynecology Department, NO36. Sanhao Street, Heping district, Shenyang, China.
| |
Collapse
|
21
|
Manzo Margiotta F, Fidanzi C, Pardossi S, Michelucci A, Granieri G, Salvia G, Bevilacqua M, Morganti R, Panduri S, Bagnoni G, Romanelli M, Janowska A, Dini V. Correlation between skin cancer and therapy with anti-IL monoclonal antibodies in patients affected by moderate-to-severe psoriasis: an observational monocentric study. Eur J Cancer Prev 2025:00008469-990000000-00200. [PMID: 39786540 DOI: 10.1097/cej.0000000000000948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Our study aimed to investigate the correlation between skin cancer and anti-interleukin (IL) therapy in patients with moderate-to-severe psoriasis. This was an observational monocentric study in which we enrolled a total of 235 patients in which 127 patients were affected by moderate-to-severe psoriasis and treated with anti-IL monoclonal antibodies (mAbs) for at least 6 months, whereas 108 patients affected by mild psoriasis were treated with topical therapies. Afterward, we performed a dermatologic visit to all the subjects, collecting anamnestic information including risk factors for skin cancer. We examined the skin lesions on their entire body by polarized and nonpolarized dermoscopy. A total of 21 suspicious lesions in the first group and 17 in the second one were removed and histologically analyzed. Twelve (9.4%) cancerous or precancerous lesions were found in the first group: seven (5.5%) basal cell carcinoma (BCC) and five actinic keratosis. The mean time to cancer onset was identified as 22 months after the start of four therapy, with an SD of 18 months, suggesting an earlier onset with respect to the start of therapy in our population. On univariate analysis, age (P = 0.001) and age of psoriasis onset (0.009) were statistically significant. Nine of 17 were skin cancers in the second group. Our study provided real-life evidence of the percentage of patients with skin cancers during therapy with anti-IL mAbs, demonstrating a good safety profile of the investigated drugs.
Collapse
Affiliation(s)
- Flavia Manzo Margiotta
- Department of Dermatology, University of Pisa
- Interdisciplinary Center of Health Science, Sant'Anna School of Advanced Studies of Pisa, Pisa
| | - Cristian Fidanzi
- Department of Dermatology, University of Pisa
- Melanoma and Skin Cancer Unit, AVNO Tuscany, Livorno
- Massa-Carrara Hospitals, Massa Carrara
| | - Simone Pardossi
- Department of Dermatology, University of Pisa
- Interdisciplinary Center of Health Science, Sant'Anna School of Advanced Studies of Pisa, Pisa
| | - Alessandra Michelucci
- Department of Dermatology, University of Pisa
- Interdisciplinary Center of Health Science, Sant'Anna School of Advanced Studies of Pisa, Pisa
| | | | | | | | - Riccardo Morganti
- Statistical Support to Clinical Trials Department, University of Pisa, Pisa, Italy
| | | | - Giovanni Bagnoni
- Melanoma and Skin Cancer Unit, AVNO Tuscany, Livorno
- Massa-Carrara Hospitals, Massa Carrara
| | | | | | | |
Collapse
|
22
|
Lei ZY, Wu J, Zhang BH, Xiang W, Wang M, Li BB, Dai MC. High FGF18 expression levels predict poor prognosis in endometrial carcinoma patients and promote tumor growth and metastasis. J Int Med Res 2025; 53:3000605241311402. [PMID: 39852234 PMCID: PMC11760137 DOI: 10.1177/03000605241311402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/16/2024] [Indexed: 01/26/2025] Open
Abstract
OBJECTIVE To investigate if fibroblast growth factor 18 (FGF18) expression plays an important role in endometrial carcinoma (EC). METHODS The clinicopathological associations and prognostic value of FGF18 expression were retrospectively analyzed in 190 patients with EC. FGF18 expression was stably knocked down in EC cell lines. Changes in cell proliferation, migration, and invasion rates were examined via cell behavior experiments. Tumor growth was investigated using a xenograft mouse model. RNA sequencing (RNA-seq) was performed to identify differentially expressed genes (DEGs) in HEC-1-B cells after FGF18 knockdown, followed by pathway enrichment analysis of the DEGs. RESULTS High FGF18 expression levels were closely correlated with EC clinicopathological features, such as histological subtype, FIGO stage, depth of myometrial invasion, and tumor size. Moreover, EC patients with high FGF18 expression levels had poorer overall survival. FGF18 knockdown in EC cells revealed its role in promoting tumor cell proliferation, migration, and invasion in vitro, as well as tumor growth in vivo. RNA-seq of HEC-1-B cells revealed that the DEGs were enriched in signaling pathways related to cell proliferation and migration. CONCLUSIONS Overexpression of FGF18 may serve as a prognostic biomarker for EC patients and is a potential therapeutic target for treating this disease.
Collapse
Affiliation(s)
- Zheng-Yao Lei
- Department of Pathology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jian Wu
- Department of Pathology, Punan Branch of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bao-Hua Zhang
- Department of Obstetrics and Gynecology, Punan Branch of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Xiang
- Department of Pathology, Punan Branch of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Min Wang
- Department of Pathology, Punan Branch of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bin-bin Li
- Department of Pathology, Punan Branch of Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ming-Cheng Dai
- School of Clinical Medicine, Harbin Medical University, Harbin City, Heilongjiang, Province, China
| |
Collapse
|
23
|
Zhang H, Jiang J, Chen X, Zhu F, Fu F, Chen A, Fu L, Mao D. Liu-Shen-Wan inhibits PI3K/Akt and TRPV1 signaling alleviating bone cancer pain in rats. Cancer Biol Ther 2024; 25:2432098. [PMID: 39587385 PMCID: PMC11601056 DOI: 10.1080/15384047.2024.2432098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/06/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024] Open
Abstract
Patients with advanced-stage cancers often suffer from severe pain caused by bone metastasis and destruction, for which effective treatment options are limited. Liu-Shen-Wan (LSW) is a widely recognized herbal formula utilized for pain relief. This study aims to elucidate the effects of LSW on bone cancer pain (BCP). In this study, the pharmacology of LSW on BCP was screened by network pharmacology. A BCP model was conducted using Walker 256 cells. Paw withdrawal threshold and paw withdrawal latency were employed as measures to assess the pain threshold in rats. The pathways and cell types of LSW against BCP were explored. Next, the impact of LSW on Walker 256 cells was evaluated, and UPLC-MS was utilized to identify the active ingredients of LSW. Furthermore, the effects of the key active ingredient, Bufalin, on the BCP rats were evaluated. There were 275 shared targets between LSW and BCP, which were enriched in neural tissue ligand-receptor interaction pathway. LSW increased pain threshold and decreased inflammatory cytokines levels in BCP rats by inhibiting PI3K/Akt and transient receptor potential vanilloid 1 (TRPV1) signaling through astrocytes and microglia. LY294002 further alleviated BCP in rats, while the effects were reversed after treatment with insulin-like growth factor 1 (IGF-1). Both LSW and its active ingredient Bufalin were shown to inhibit the viability and migration of Walker 256 cells and induce apoptosis. Bufalin appears to be the key active ingredient of LSW and exerts its pain-relieving effects by suppressing PI3K/Akt and TRPV1 signaling in BCP.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Hainan Hospital, Haikou, Hainan, China
| | - Jingwen Jiang
- Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Hainan Hospital, Haikou, Hainan, China
| | - Xuewu Chen
- Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Hainan Hospital, Haikou, Hainan, China
| | - Fengting Zhu
- Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Hainan Hospital, Haikou, Hainan, China
| | - Fangfang Fu
- Department of Oncology, Affiliated Hainan Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Haikou, Hainan, China
| | - Aiying Chen
- Department of Oncology, Affiliated Hainan Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Haikou, Hainan, China
| | - Lei Fu
- Department of Dermatology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Hainan Hospital, Haikou, Hainan, China
| | - Dan Mao
- Department of Integrated Traditional Chinese and Western Medicine, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
24
|
Luo A, Liu H, Huang C, Wei S. Exosome-transmitted circular RNA circ-LMO7 facilitates the progression of osteosarcoma by regulating miR-21-5p/ARHGAP24 axis. Cancer Biol Ther 2024; 25:2343450. [PMID: 38742566 PMCID: PMC11095575 DOI: 10.1080/15384047.2024.2343450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 04/11/2024] [Indexed: 05/16/2024] Open
Abstract
The potential function and mechanism of circRNAs in regulating malignant performances of Osteosarcoma (OS) cells have not been well investigated. The expression level of CircLMO7, miR-21-5p and ARHGAP24 were detected by RT-qPCR. The relationship between miR-21-5p and circ-LMO7, as well as between miR-21-5p and ARHGAP24, was predicted and examined through bioinformatics analysis and luciferase reporter gene experiments. Moreover, OS cell growth, invasion, migration, and apoptosis were detected using the cell counting kit-8 (CCK-8), transwell and flow cytometry assays, respectively. ARHGAP24 protein level was measured using western blotting. In present study, we choose to investigate the role and mechanism of circ-LOM7 on OS cell proliferation, migration and invasion. circ-LOM7 was found to be down-regulated in OS tissues and cell lines. Enforced expression of circ-LOM7 suppressed the growth, invasion, and migration of OS cells. In contrast, decreasing circ-LMO7 expression had opposite effects. Furthermore, miR-21-5p was predicted to be sponged by circ-LMO7, and had an opposite role of circ-LMO7 in OS. Moreover, ARHGAP24 served as miR-21-5p's downstream target. Mechanistically, circ-LMO7 was packed in exosomes and acted as a cancer-suppresser on OS by sponging miR-21-5p and upregulating the expression of ARHGAP24. The exosomal circ-LMO7 expression was significantly decreased in OS cell exosomes, and co-culture experiments showed that exosomal circ-LMO7 suppressed the proliferation ability of OS cells. Circ-LMO7 exerts as a tumor suppressor in OS, and the circ-LMO7/miR-21-5P/ARHGAP24 axis is involved in OS progression.
Collapse
Affiliation(s)
- Anyu Luo
- Department of Orthopedics, Hanyang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Hanlin Liu
- Department of Orthopedics, Hanyang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Chen Huang
- Department of Orthopedics, Hanyang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Sheng Wei
- Department of Orthopedics, Hanyang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
25
|
Li W, Zhang Q, Ni M, Li B, Chen Z, Shen Q, Lin Z, Cheng C, Yao D, Qi S, Ding X, Shen H, Liu X, Tang Z, Huang X, Zhao J, Liu Z. Upregulated YTHDC1 mediates trophoblastic dysfunction inducing preterm birth in ART conceptions through enhanced RPL37 translation. Cell Mol Life Sci 2024; 82:17. [PMID: 39725796 DOI: 10.1007/s00018-024-05467-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/01/2024] [Accepted: 10/01/2024] [Indexed: 12/28/2024]
Abstract
Assisted reproductive technology (ART) pregnancies present a higher risk of singleton preterm birth than natural pregnancies, but the underlying molecular mechanism remains largely unknown. RNA m6A modification is a key epigenetic mechanism regulating cellular function, but the role of m6A modification, especially its "reader" YTHDC1, in preterm delivery remains undefined. To delineate the role and epigenetic mechanism of m6A modification in ART preterm delivery, the effects of YTHDC1 on trophoblastic function were evaluated by CCK-8, EdU, Transwell, and flow cytometry analyses post its overexpression or knockdown. Downstream signaling pathways of YTHDC1 were investigated by RNA-seq, and targeted mRNAs were explored by RIP-seq and MeRIP-seq. Upstream transcriptional factors of YTHDC1 were determined by ChIP-seq and luciferase reporter assays. Elevated YTHDC1 was detected in human ART-conceived preterm placentas and in murine preterm placentas post estradiol (E2) exposure. In vitro experiments showed that YTHDC1 promoted trophoblastic cell proliferation and migration, but inhibited cell apoptosis. Mechanistically, E2 was proven to upregulate YTHDC1 expression via retinoid X receptor alpha (RXRA) in trophoblastic cells. Enhanced YTHDC1 expression augmented the translation of RPL37 in an m6A-dependent manner by binding to m6A-modified RPL37 mRNA and concomitantly promoted the overall translational output. Importantly, administration of siRNA targeting YTHDC1 effectively delayed the progression of preterm delivery. In conclusion, the identified E2/RXRA/YTHDC1/RPL37 axis provides new insights into the epigenetic mechanism underlying ART-associated preterm delivery. The findings offer a potential prognostic biomarker and therapeutic target for preterm delivery.
Collapse
Affiliation(s)
- Wei Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Qianqian Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Meng Ni
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Baihe Li
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Ze Chen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Qianwen Shen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Zhenying Lin
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Chunyu Cheng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Dongting Yao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Sudong Qi
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xiya Ding
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Haiqing Shen
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
| | - Xiaorui Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Zheng Tang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
| | - Xiaoyi Huang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China
| | - Jiuru Zhao
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| | - Zhiwei Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, 910# Hengshan Road, Shanghai, China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China.
| |
Collapse
|
26
|
Wang J, Zhang H, Li J, Ni X, Yan W, Chen Y, Shi T. Exosome-derived proteins in gastric cancer progression, drug resistance, and immune response. Cell Mol Biol Lett 2024; 29:157. [PMID: 39719600 PMCID: PMC11667977 DOI: 10.1186/s11658-024-00676-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024] Open
Abstract
Gastric cancer (GC) represents a prevalent malignancy globally, often diagnosed at advanced stages owing to subtle early symptoms, resulting in a poor prognosis. Exosomes are extracellular nano-sized vesicles and are secreted by various cells. Mounting evidence indicates that exosomes contain a wide range of molecules, such as DNA, RNA, lipids, and proteins, and play crucial roles in multiple cancers including GC. Recently, with the rapid development of mass spectrometry-based detection technology, researchers have paid increasing attention to exosomal cargo proteins. In this review, we discussed the origin of exosomes and the diagnostic and prognostic roles of exosomal proteins in GC. Moreover, we summarized the biological functions of exosomal proteins in GC processes, such as proliferation, metastasis, drug resistance, stemness, immune response, angiogenesis, and traditional Chinese medicine therapy. In summary, this review synthesizes current advancements in exosomal proteins associated with GC, offering insights that could pave the way for novel diagnostic and therapeutic strategies for GC in the foreseeable future.
Collapse
Affiliation(s)
- Jiayu Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huan Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
| | - Juntao Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiangyu Ni
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenying Yan
- Department of Bioinformatics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China.
- Center for Systems Biology, Soochow University, Suzhou, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou, China.
| | - Yueqiu Chen
- Department of Cardiovascular Surgery of The First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215007, China.
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
27
|
Li D, Chu X, Ma Y, Zhang F, Tian X, Yang Y, Yang Y. Tumor-derived exosomes: Unravelling the pathogenesis of pancreatic cancer with liver metastases and exploring the potential for clinical translation. Cancer Lett 2024; 611:217403. [PMID: 39709178 DOI: 10.1016/j.canlet.2024.217403] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Pancreatic cancer (PC) is one of the most malignant solid cancers, and PC metastasis, particularly liver metastasis, is a major cause of cancer mortality. A key event in tumor metastasis is the formation of pre-metastatic niche (PMN), which provides a microenvironment conducive to tumor cells colonization and progression. Various molecules loaded in tumor-derived exosomes (TDEs) contribute to PMN formation and distant tumor metastasis, by regulating immune and stromal cell function, inducing angiogenesis, and promoting metabolic reprogramming. Therefore, therapies targeting PMN may offer novel advantages to prevent tumor metastasis at an earlier stage. In this review, we summarize multifaceted mechanisms underlying hepatic PMN formation, with a focus on how PC TDEs participate in angiogenesis and vascular permeability, create immune suppressive microenvironment, remodel the extracellular matrix, and regulate metabolic reprogramming. In addition, we highlight the promise of TDEs for early diagnosis and effective therapy of PC liver metastases.
Collapse
Affiliation(s)
- Dongqi Li
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xiangyu Chu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, State Key Lab of Digestive Health, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yongsu Ma
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Fusheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xiaodong Tian
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China.
| | - Yanlian Yang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
28
|
Li J, Zhou W, Wang H, Huang M, Deng H. Exosomal circular RNAs in tumor microenvironment: An emphasis on signaling pathways and clinical opportunities. MedComm (Beijing) 2024; 5:e70019. [PMID: 39584047 PMCID: PMC11586091 DOI: 10.1002/mco2.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/26/2024] Open
Abstract
Exosomes can regulate the malignant progression of tumors by carrying a variety of genetic information and transmitting it to target cells. Recent studies indicate that exosomal circular RNAs (circRNAs) regulate multiple biological processes in carcinogenesis, such as tumor growth, metastasis, epithelial-mesenchymal transition, drug resistance, autophagy, metabolism, angiogenesis, and immune escape. In the tumor microenvironment (TME), exosomal circRNAs can be transferred among tumor cells, endothelial cells, cancer-associated fibroblasts, immune cells, and microbiota, affecting tumor initiation and progression. Due to the high stability and widespread presence of exosomal circRNAs, they hold promise as biomarkers for tumor diagnosis and prognosis prediction in blood and urine. In addition, designing nanoparticles targeting exosomal circRNAs and utilizing exosomal circRNAs derived from immune cells or stem cells provide new strategies for cancer therapy. In this review, we examined the crucial role of exosomal circRNAs in regulating tumor-related signaling pathways and summarized the transmission of exosomal circRNAs between various types of cells and their impact on the TME. Finally, our review highlights the potential of exosomal circRNAs as diagnostic and prognostic prediction biomarkers, as well as suggesting new strategies for clinical therapy.
Collapse
Affiliation(s)
- Junshu Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Wencheng Zhou
- Department of Medical AestheticsWest China School of Public Health and West China Fourth HospitalSichuan UniversityChengduChina
| | - Huiling Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Meijuan Huang
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical OncologyCancer CenterWest China Hospital, Sichuan UniversityChengduChina
| | - Hongxin Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
29
|
Wang MH, Liu ZH, Zhang HX, Liu HC, Ma LH. Hsa_circRNA_000166 accelerates breast cancer progression via the regulation of the miR-326/ELK1 and miR-330-5p/ELK1 axes. Ann Med 2024; 56:2424515. [PMID: 39529543 PMCID: PMC11559033 DOI: 10.1080/07853890.2024.2424515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSES To probe the expression, clinical significance, roles, and molecular mechanisms of circRNA_000166 in breast cancer (BC). METHODS Clinical tissue samples were gathered from 84 BC patients who underwent surgery at the Affiliated Hospital of Chengde Medical College. Clinical data were obtained from medical records and postoperative follow-up. Expression levels of circRNA_000166, miR-326, miR-330-5p, and ELK1 mRNA in BC tissues and cells were measured by qRT-PCR, and ELK1 protein levels were assessed by WB. Pearson's correlation analysis evaluated the interrelationships between these RNAs in clinical samples. Luciferase reporter assays verified the interactions between miR-326/miR-330-5p and circRNA_000166, as well as between miR-326/miR-330-5p and ELK1. Cell proliferation, migration, and apoptosis were examined using CCK-8, colony formation, transwell, and flow cytometry assays, respectively. RESULTS CircRNA_000166 was highly expressed in BC tissues and inversely correlated with miR-326/miR-330-5p levels but positively with ELK1 mRNA levels. ELK1 mRNA also inversely associated with miR-326/miR-330-5p levels in BC tissues. Importantly, our findings demonstrated that circRNA_000166 targets miR-326 and miR-330-5p, while ELK1 is the target of miR-326 and miR-330-5p in BC cells. CircRNA_000166 levels positively correlated with tumour size, TNM stage, histological grade, and lymph node metastasis, and negatively associated with postoperative progression-free survival (PFS) and overall survival (OS) in BC patients. CircRNA_000166 was also highly expressed in BC cells, and knockdown of circRNA_000166 reduced proliferation and migration, and increased apoptosis via miR-326/ELK1 and miR-330-5p/ELK1 pathways in vitro. CONCLUSION CircRNA_000166 enhances BC progression through miR-326/ELK1 and miR-330-5p/ELK1 pathways and shows potential as a biomarker for BC diagnosis and treatment.
Collapse
Affiliation(s)
- Ming-Hui Wang
- Department of Breast Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| | - Zi-Hui Liu
- Department of Pathology, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| | - Hong-Xu Zhang
- Department of Breast Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| | - Han-Cheng Liu
- Department of Breast Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| | - Li-Hui Ma
- Department of Breast Surgery, Affiliated Hospital of Chengde Medical University, Chengde, Hebei, China
| |
Collapse
|
30
|
Jiang A, Liu Y, He Z, Liu W, Yang Q, Fang Y, Zhu B, Wu X, Ye H, Ye B, Gao S, Qu L, Xu W, Luo P, Wang L. TDERS, an exosome RNA-derived signature predicts prognosis and immunotherapeutic response in clear cell renal cell cancer: a multicohort study. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:382-394. [PMID: 39735439 PMCID: PMC11674438 DOI: 10.1016/j.jncc.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 06/08/2024] [Accepted: 07/23/2024] [Indexed: 12/31/2024] Open
Abstract
Background Tumor-derived exosomes are involved in tumor progression and immune invasion and might function as promising noninvasive approaches for clinical management. However, there are few reports on exosom-based markers for predicting the progression and adjuvant therapy response rate among patients with clear cell renal cell carcinoma (ccRCC). Methods The signatures differentially expressed in exosomes from tumor and normal tissues from ccRCC patients were correspondingly deregulated in ccRCC tissues. We adopted a two-step strategy, including Lasso and bootstrapping, to construct a novel risk stratification system termed the TDERS (Tumor-Derived Exosome-Related Risk Score). During the testing and validation phases, we leveraged multiple external datasets containing over 2000 RCC cases from eight cohorts and one inhouse cohort to evaluate the accuracy of the TDERS. In addition, enrichment analysis, immune infiltration signatures, mutation landscape and therapy sensitivity between the high and low TDERS groups were compared. Finally, the impact of TDERS on the tumor microenvironment (TME) was also analysed in our single-cell datasets. Results TDERS consisted of 12 mRNAs deregulated in both exosomes and tissues from patients with ccRCC. TDERS achieved satisfactory performance in both prognosis and immune checkpoint inhibitor (ICI) response across all ccRCC cohorts and other pathological types, since the average area under the curve (AUC) to predict 5-year overall survival (OS) was larger than 0.8 across the four cohorts. Patients in the TDERS high group were resistant to ICIs, while mercaptopurine might function as a promising agent for those patients. Patients with a high TDERS were characterized by coagulation and hypoxia, which induced hampered tumor antigen presentation and relative resistance to ICIs. In addition, single cells from 12 advanced samples validated this phenomenon since the interaction between dendritic cells and macrophages was limited. Finally, PLOD2, which is highly expressed in fibro- and epi‑tissue, could be a potential therapeutic target for ccRCC patients since inhibiting PLOD2 altered the malignant phenotype of ccRCC in vitro. Conclusion As a novel, non-invasive, and repeatable monitoring tool, the TDERS could work as a robust risk stratification system for patients with ccRCC and precisely inform treatment decisions about ICI therapy.
Collapse
Affiliation(s)
- Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ziwei He
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qiwei Yang
- Department of Urology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Urology, the Third Affiliated Hospital of Naval Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Baohua Zhu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xiaofeng Wu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Huamao Ye
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Bicheng Ye
- School of Clinical Medicine, Medical College of Yangzhou Polytechnic College, Yangzhou, China
| | - Shunxiang Gao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, and Human Phenome Institute, Fudan University, Shanghai, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
31
|
Li X, Wu Y, Jin Y. Exosomal LncRNAs and CircRNAs in lung cancer: Emerging regulators and potential therapeutic targets. Noncoding RNA Res 2024; 9:1069-1079. [PMID: 39022675 PMCID: PMC11254510 DOI: 10.1016/j.ncrna.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Lung cancer remains one of the most prevalent and lethal malignancies globally, characterized by high incidence and mortality rates among all cancers. The delayed diagnosis of lung cancer at intermediate to advanced stages frequently leads to suboptimal treatment outcomes. To improve the management of this disease, it is imperative to identify new, highly sensitive prognostic and diagnostic biomarkers. Exosomes, extracellular vesicles with a lipid-bilayer structure and a size range of 30-150 nm, are pivotal in intercellular communication and play significant roles in lung cancer progression. Non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are highly prevalent within exosomes and play a crucial role in various pathophysiological processes mediated by these extracellular vesicles. Beyond their established functions in miRNA and protein sequestration, these ncRNAs are involved in regulating translation and interactions within exosomes. Numerous studies have highlighted the importance of exosomal lncRNAs and circRNAs in influencing epithelial-mesenchymal transition (EMT), angiogenesis, proliferation, invasion, migration, and metastasis in lung cancer. Due to their unique functional characteristics, these molecules are promising therapeutic targets and biomarkers for diagnosis and prognosis. This review provides a succinct summary of the formation of exosomal lncRNAs and circRNAs, clarifies their biological roles, and thoroughly explains the mechanisms by which they participate in the progression of lung cancer. Finally, we discuss the potential clinical applications and challenges associated with exosomal lncRNAs and circRNAs in lung cancer.
Collapse
Affiliation(s)
- Xia Li
- Center of Molecular Diagnostic, Northern Jiangsu People's Hospital of Jiangsu Province, Yangzhou, 225001, China
| | - Yunbing Wu
- Department of Medicine Laboratory, Northern Jiangsu People's Hospital of Jiangsu Province, Yangzhou, 225001, China
| | - Yue Jin
- Center of Molecular Diagnostic, Northern Jiangsu People's Hospital of Jiangsu Province, Yangzhou, 225001, China
| |
Collapse
|
32
|
Zhang J, Luo X, Guo C, Dai Z, Tang X, Zhang F, Jiao Q, Lin S, Zou L, Zhang Z, Lv XB. LncRNA GClnc1 promotes osteosarcoma progression by stabilizing NONO and blocking FBXW7-mediated ubiquitination. BMC Cancer 2024; 24:1375. [PMID: 39523321 PMCID: PMC11552323 DOI: 10.1186/s12885-024-13138-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) plays a vital role in the occurrence and development of varieties of tumors. Previous studies have shown that lncRNA GClnc1 is highly expressed in osteosarcoma (OS). However, the mechanism of lncRNA GClnc1 in osteosarcoma has not been fully elucidated. In this study, we investigated the biological roles of lncRNA GClnc1 in osteosarcoma and unveiled its underlying mechanisms. METHODS The expression of lncRNA GClnc1 in OS cells was detected by real-time quantitative PCR (qRT-PCR). The functional roles of lncRNA GClnc1 were examined by CCK8, trans-well, scratch wound healing assay, colony formation, and apoptosis assays in osteosarcoma cells upon silencing or overexpressing GClnc1. Western blot analysis, qRT-PCR, and RNA co-immunoprecipitation (RIP) assays were used to detect the interaction between lncRNA GClnc1 and NONO. RESULTS The expression of lncRNA GClnc1 was up-regulated in osteosarcoma cell lines. Knockdown of lncRNA GClnc1 suppressed the cell growth, migration, and invasion of OS cells, whereas the over-expression of GClnc1 improved the proliferation, migration, and invasion of OS cells. Mechanistically, we identified that lncRNA GClnc1 regulates the stability of NONO by blocking FBXW7-mediated ubiquitination degradation. Additionally, overexpression of NONO can reverse GClnc1 silencing exerted suppression of the cell proliferation, migration, and invasion, and vice versa. CONCLUSIONS Our study elucidated that lncRNA GClnc1 participates in the progression of OS by regulating the NONO signal pathway. Targeting GClnc1 provides a potential target for future clinical treatment of OS.
Collapse
Affiliation(s)
- Jiongfeng Zhang
- Jiangxi Key Laboratory of Oncology, The Central Lab of The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, China
- Department of Orthopedics, Nanchang Key Laboratory of Orthopaedics, The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330008, China
| | - Xiaohui Luo
- Jiangxi Key Laboratory of Oncology, The Central Lab of The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, China
- Department of Orthopedics, Nanchang Key Laboratory of Orthopaedics, The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330008, China
| | - Chong Guo
- Jiangxi Key Laboratory of Oncology, The Central Lab of The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, China
- Department of Orthopedics, Nanchang Key Laboratory of Orthopaedics, The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330008, China
| | - Zhengzai Dai
- Jiangxi Key Laboratory of Oncology, The Central Lab of The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, China
| | - Xiaofeng Tang
- Department of Orthopedics, Nanchang Key Laboratory of Orthopaedics, The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330008, China
| | - Feifei Zhang
- Department of Orthopedics, Nanchang Key Laboratory of Orthopaedics, The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330008, China
| | - Quanhui Jiao
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shifan Lin
- Jiangxi Key Laboratory of Oncology, The Central Lab of The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, China
| | - Le Zou
- Jiangxi Key Laboratory of Oncology, The Central Lab of The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, China
| | - Zhiping Zhang
- Jiangxi Key Laboratory of Oncology, The Central Lab of The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, North 128 Xiangshan Road, Nanchang, 330008, China.
| | - Xiao-Bin Lv
- Department of Orthopedics, Nanchang Key Laboratory of Orthopaedics, The First Hospital of Nanchang, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330008, China.
| |
Collapse
|
33
|
Li Q, Zhao N, Ding X, Zhao J. METTL14-mediated m6A modification upregulates HOXB13 expression to activate NF-κB and exacerbate cervical cancer progression. Mol Cell Oncol 2024; 11:2423986. [PMID: 39534063 PMCID: PMC11556271 DOI: 10.1080/23723556.2024.2423986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/13/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Cervical cancer (CC) is one of the common malignant tumors in women, and the incidence rate is located in the second place of female tumors. As a major RNA N6-methyladenosine (m6A) methyltransferase, methyltransferase-like 14 (METTL14) is involved in tumor progression by catalyzing methylation modifications in mRNAs. However, the molecular mechanism of METTL14-mediated m6A modification in CC remains not fully revealed. The expression of METTL14 was detected by RT-qPCR and western blot. Cell function was assayed by cell counting kit-8 (CCK-8) assay and flow cytometry analysis. Methylated RNA immunoprecipitation (MeRIP) was used to confirm the relationship between METTL14 and homeobox B13 (HOXB13). In our study, we found that the level of METTL14 was elevated in CC tissues and cells compared with their controls. The inhibition of METTL14 significantly impaired cell proliferation and the epithelial-mesenchymal transition (EMT) process, while also induced apoptosis in HeLa and C33A cells. Furthermore, our findings indicated that homeobox B13 (HOXB13) was a target of METTL14, which positively regulated the expression of HOXB13 in an m6A-dependent manner. Rescue experiments indicated that overexpression of HOXB13 effectively reversed the tumor suppression induced by METTL14 knockdown. Finally, we confirmed that METTL14-modified HOXB13 exerted an oncogenic effect through activation of the nuclear factor kappa B (NF-κB) pathway. In conclusion, our data demonstrated that the m6A modification of HOXB13, mediated by METTL14, facilitated the advancement of CC through targeting the NF-κB pathway, which may be a potential molecular target for the treatment of CC.
Collapse
Affiliation(s)
- Qian Li
- Department of Obstetrics and Gynecology, 926th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Kaiyuan, China
| | - Na Zhao
- Department of Obstetrics and Gynecology, 926th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Kaiyuan, China
| | - Xuejing Ding
- Department of Obstetrics and Gynecology, 926th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Kaiyuan, China
| | - Jufen Zhao
- Department of Obstetrics and Gynecology, 926th Hospital of the Joint Logistics Support Force of the Chinese People’s Liberation Army, Kaiyuan, China
| |
Collapse
|
34
|
Pan W, Miao Q, Yin W, Li X, Ye W, Zhang D, Deng L, Zhang J, Chen M. The role and clinical applications of exosomes in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:43. [PMID: 39624083 PMCID: PMC11609145 DOI: 10.20517/cdr.2024.97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/27/2024] [Accepted: 10/16/2024] [Indexed: 01/03/2025]
Abstract
Tumor-secreted exosomes are heterogeneous multi-signal messengers that support cancer growth and dissemination by mediating intercellular crosstalk and activating signaling pathways. Distinct from previous reviews, we focus intently on exosome-therapeutic resistance dynamics and summarize the new findings about the regulation of cancer treatment resistance by exosomes, shedding light on the complex processes via which these nanovesicles facilitate therapeutic refractoriness across various malignancies. Future research in exosome biology can potentially transform diagnostic paradigms and therapeutic interventions for cancer management. This review synthesizes recent insights into the exosome-driven regulation of cancer drug resistance, illuminates the sophisticated mechanisms by which these nanovesicles facilitate therapeutic refractoriness across various malignancies, and summarizes some strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Wenxuan Pan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, China
- Authors contributed equally
| | - Qun Miao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, China
- Authors contributed equally
| | - Wenqian Yin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, China
| | - Xiaobo Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, China
| | - Wencai Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, China
| | - Dongmei Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, China
| | - Lijuan Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, Guangdong, China
| | - Junqiu Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, China
| | - Minfeng Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, Guangdong, China
- College of Pharmacy, Jinan University, Guangzhou 510632, Guangdong, China
| |
Collapse
|
35
|
Zou Y, Yang A, Chen B, Deng X, Xie J, Dai D, Zhang J, Tang H, Wu T, Zhou Z, Xie X, Wang J. crVDAC3 alleviates ferroptosis by impeding HSPB1 ubiquitination and confers trastuzumab deruxtecan resistance in HER2-low breast cancer. Drug Resist Updat 2024; 77:101126. [PMID: 39243601 DOI: 10.1016/j.drup.2024.101126] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 07/24/2024] [Accepted: 08/05/2024] [Indexed: 09/09/2024]
Abstract
AIMS With the wide application of trastuzumab deruxtecan (T-DXd), the survival of HER2-low breast cancer patients is dramatically improved. However, resistance to T-DXd still exists in a subset of patients, and the molecular mechanism remains unclear. METHODS An in vivo shRNA lentiviral library functional screening was performed to identify potential circular RNA (crRNA) that mediates T-DXd resistance. RNA pull-down, mass spectrometry, RNA immunoprecipitation, and co-immunoprecipitation assays were conducted to investigate the molecular mechanism. Ferroptosis was detected using C11-BODIPY, Liperfluo, FerroOrange staining, glutathione quantification, malondialdehyde quantification, and transmission electron microscopy. Molecular docking, virtual screening, and patient-derived xenograft (PDX) models were used to validate therapeutic agents. RESULTS VDAC3-derived crRNA (crVDAC3) ranked first in functional shRNA library screening. Knockdown of crVDAC3 increased the sensitivity of HER2-low breast cancer cells to T-DXd treatment. Further mechanistic research revealed that crVDAC3 specifically binds to HSPB1 protein and inhibits its ubiquitination degradation, leading to intracellular accumulation and increased levels of HSPB1 protein. Notably, suppression of crVDAC3 dramatically increases excessive ROS levels and labile iron pool accumulation. Inhibition of crVDAC3 induces ferroptosis in breast cancer cells by reducing HSPB1 expression, thereby mediating T-DXd resistance. Through virtual screening and experimental validation, we identified that paritaprevir could effectively bind to crVDAC3 and prevent its interaction with HSPB1 protein, thereby increasing ubiquitination degradation of HSPB1 protein to overcome T-DXd resistance. Finally, we validated the enhanced therapeutic efficacy of T-DXd by paritaprevir in a HER2-low PDX model. CONCLUSION This finding reveals the molecular mechanisms underlying T-DXd resistance in HER2-low breast cancer. Our study provides a new strategy to overcome T-DXd resistance by inhibiting the interaction between crVDAC3 and HSPB1 protein.
Collapse
Affiliation(s)
- Yutian Zou
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Anli Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Bo Chen
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xinpei Deng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jindong Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Danian Dai
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jinhui Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Tao Wu
- Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, China.
| | - Zhigang Zhou
- Changde Hospital, Xiangya School of Medicine, Central South University (The first people's hospital of Changde city), Changde, China.
| | - Xiaoming Xie
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Jin Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
36
|
Heidari N, Vosough M, Bagherifard A, Sami SH, Sarabi PA, Behmanesh A, Shams R. Exploring circulating MiRNA signature for osteosarcoma detection: Bioinformatics-based analyzing and validation. Pathol Res Pract 2024; 263:155615. [PMID: 39378797 DOI: 10.1016/j.prp.2024.155615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/15/2024] [Accepted: 09/25/2024] [Indexed: 10/10/2024]
Abstract
Early detection followed by efficient treatment still remain a considerable challenge for osteosarcoma (OS), indicating the importance of emerging innovative diagnostic methods. Circulating miRNAs offer a promising and non-invasive approach to assess the OS molecular landscapes. This study utilized RNAseq data from OS plasma miRNA expression profiles (PRJEB30542) and PCR Array data (GSE65071) from GEO and ENA databases. In total, 43 miRNAs demonstrated significant differential expression in OS samples of training dataset. A diagnostic model, including hsa-miR-30a-5p, hsa-miR-556-3p, hsa-miR-200a-3p, and hsa-miR-582-5p was identified through multivariate logistic regression analysis and demonstrated significant efficacy in differentiating OS patients from healthy controls in the validation group (AUC: 0.917, sensitivity: 1, specificity: 0.85). The result of target gene prediction and functional enrichment analyses revealed significant associations with terms such as epithelial morphogenesis, P53 and Wnt signaling pathways, and neoplasm metastasis. Further bioinformatics-based evaluations showed that the down-regulation of these miRNAs significantly correlates with poor prognosis and lower survival rate in OS patients and propose their tumor suppressor function in pathogenesis of OS. Furthermore, the study developed a miRNA-mRNA subnetwork that connects these miRNAs to the P53 and Wnt signaling pathways, which are critical pathways with oncogenic effects on OS progression. This comprehensive approach not only presents a promising diagnostic model but also proposes potential molecular markers for OS early diagnosis, making prognosis, and targeted therapy. The identified miRNA-mRNA functional axis holds promise as a valuable resource for further research in understanding OS pathogenesis and establishing therapeutic modalities.
Collapse
Affiliation(s)
- Negar Heidari
- Department of Cellular and Molecular Biology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| | - Abolfazl Bagherifard
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Sam Hajialilo Sami
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Pedram Asadi Sarabi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Ali Behmanesh
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Roshanak Shams
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Zhou Q, Zhou L, Chen X, Chen Q, Hao L. Crosstalk Between the Intratumoral Microbiota and the Tumor Microenvironment: New Frontiers in Solid Tumor Progression and Treatment. Cancer Rep (Hoboken) 2024; 7:e70063. [PMID: 39559964 PMCID: PMC11574561 DOI: 10.1002/cnr2.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 10/06/2024] [Accepted: 10/29/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND The microbiota plays a significant role in the tumor microenvironment, and its impact on tumor development and treatment outcome cannot be overlooked. Thus, it is essential to comprehend the interactions between the microbiota and the tumor microenvironment. RECENT FINDINGS With the advent of next-generation sequencing, microbiota research has advanced significantly in recent years. The interaction between the intratumoral microbiota and the tumor microenvironment is an emerging area of research that holds great promise for understanding and treating solid tumor progression. This crosstalk between the intratumoral microbiota and the tumor microenvironment is a complex process that involves a multitude of factors, including the immune system, cellular signaling pathways, and metabolic processes. The origin of the intratumoral microbiota differs between various solid tumor, and the quantity and diversity of intratumoral microbiota also fluctuate significantly within each solid tumor. CONCLUSION The aim of this review is to provide a detailed summary of the intratumoral microbiota in various types of solid tumors. This will include an analysis of their origins, differences, and how they impact the progression of solid tumors. Furthermore, we will emphasize the significant potential that the intratumoral microbiota holds for the diagnosis and treatment of solid tumors.
Collapse
Affiliation(s)
- Qing Zhou
- Central Laboratory, The People's Hospital of Baoan Shenzhen, Shenzhen, China
| | - Lijun Zhou
- Department of Urology, The People's Hospital of Baoan Shenzhen, Shenzhen, China
| | - Xi Chen
- Central Laboratory, The People's Hospital of Baoan Shenzhen, Shenzhen, China
| | - Qiuyan Chen
- Science and Education Department, Shenzhen Baoan Shiyan People's Hospital, Shenzhen, China
| | - Lu Hao
- Science and Education Department, Shenzhen Baoan Shiyan People's Hospital, Shenzhen, China
| |
Collapse
|
38
|
Dong D, Yu X, Xu J, Yu N, Liu Z, Sun Y. Cellular and molecular mechanisms of gastrointestinal cancer liver metastases and drug resistance. Drug Resist Updat 2024; 77:101125. [PMID: 39173439 DOI: 10.1016/j.drup.2024.101125] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/24/2024]
Abstract
Distant metastases and drug resistance account for poor survival of patients with gastrointestinal (GI) malignancies such as gastric cancer, pancreatic cancer, and colorectal cancer. GI cancers most commonly metastasize to the liver, which provides a unique immunosuppressive tumour microenvironment to support the development of a premetastatic niche for tumor cell colonization and metastatic outgrowth. Metastatic tumors often exhibit greater resistance to drugs than primary tumors, posing extra challenges in treatment. The liver metastases and drug resistance of GI cancers are regulated by complex, intertwined, and tumor-dependent cellular and molecular mechanisms that influence tumor cell behavior (e.g. epithelial-to-mesenchymal transition, or EMT), tumor microenvironment (TME) (e.g. the extracellular matrix, cancer-associated fibroblasts, and tumor-infiltrating immune cells), tumor cell-TME interactions (e.g. through cytokines and exosomes), liver microenvironment (e.g. hepatic stellate cells and macrophages), and the route and mechanism of tumor cell dissemination (e.g. circulating tumor cells). This review provides an overview of recent advances in the research on cellular and molecular mechanisms that regulate liver metastases and drug resistance of GI cancers. We also discuss recent advances in the development of mechanism-based therapy for these GI cancers. Targeting these cellular and molecular mechanisms, either alone or in combination, may potentially provide novel approaches to treat metastatic GI malignancies.
Collapse
Affiliation(s)
- Daosong Dong
- Department of Pain, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xue Yu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Key Laboratory of Molecular Pathology and Epidemiology of Gastric Cancer in the Universities of Liaoning Province, Shenyang, Liaoning 110001, China
| | - Jingjing Xu
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Na Yu
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Disease, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Yanbin Sun
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
39
|
Fang L, Zhu Z, Han M, Li S, Kong X, Yang L. Unlocking the potential of extracellular vesicle circRNAs in breast cancer: From molecular mechanisms to therapeutic horizons. Biomed Pharmacother 2024; 180:117480. [PMID: 39357330 DOI: 10.1016/j.biopha.2024.117480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Breast cancer remains the leading cause of cancer-related morbidity and mortality among women worldwide, underscoring the urgent need for novel diagnostic and therapeutic strategies. This review explores the emerging roles of circular RNAs (circRNAs) within extracellular vesicles (exosomes) in breast cancer. circRNAs, known for their stability and tissue-specific expression, are aberrantly expressed in breast cancer and regulate critical cellular processes such as proliferation, migration, and apoptosis, positioning them as promising biomarkers. Exosomes facilitate intercellular communication by delivering circRNAs, reflecting the physiological and pathological state of their source cells. This review highlights the multifaceted roles of exosomal circRNAs in promoting tumor growth, metastasis, and drug resistance through their modulation of tumor metabolism, the tumor microenvironment, and immune responses. In particular, we emphasize their contributions to chemotherapy resistance and their potential as both diagnostic markers and therapeutic targets. By synthesizing current research, this review provides novel insights into the clinical applications of exosomal circRNAs, offering a foundation for future studies aimed at improving breast cancer management through non-invasive diagnostics and targeted therapies.
Collapse
Affiliation(s)
- Lijuan Fang
- Department of Laboratory Medicine, Hangzhou Ninth People's Hospital, Hangzhou, Zhejaing Province 311200, China
| | - Zehua Zhu
- Department of Laboratory Medicine, Hangzhou Ninth People's Hospital, Hangzhou, Zhejaing Province 311200, China
| | - Mingyue Han
- Department of Laboratory Medicine, Hangzhou Ninth People's Hospital, Hangzhou, Zhejaing Province 311200, China
| | - Shaojie Li
- Department of Laboratory Medicine, Hangzhou Ninth People's Hospital, Hangzhou, Zhejaing Province 311200, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lusen Yang
- Department of Laboratory Medicine, Hangzhou Ninth People's Hospital, Hangzhou, Zhejaing Province 311200, China.
| |
Collapse
|
40
|
Liu Z, Cheng L, Cao W, Shen C, Qiu Y, Li C, Xiong Y, Yang SB, Chen Z, Yin X, Zhang X. Present and future use of exosomes containing proteins and RNAs in neurodegenerative diseases for synaptic function regulation: A comprehensive review. Int J Biol Macromol 2024; 280:135826. [PMID: 39322147 DOI: 10.1016/j.ijbiomac.2024.135826] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/18/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Neurodegenerative diseases (NDDs) are increasingly prevalent with global aging, demanding effective treatments. Exosomes, which contain biological macromolecules such as RNA (including miRNAs) and proteins like α-synuclein, tau, and amyloid-beta, are gaining attention as innovative therapeutics. This comprehensive review systematically explores the potential roles of exosomes in NDDs, with a particular focus on their role in synaptic dysfunction. We present the synaptic pathophysiology of NDDs and discuss the mechanisms of exosome formation, secretion, and action. Subsequently, we review the roles of exosomes in different types of NDDs, such as Alzheimer's disease and Parkinson's disease, with a special focus on their regulation of synaptic function. In addition, we explore the potential use of exosomes as biomarkers, as well as the challenges and opportunities in their clinical application. We provide perspectives on future research directions and development trends to provide a more comprehensive understanding of and guidance for the application of exosomes in the treatment of NDDs. In conclusion, exosomes rich in biological macromolecules, as a novel therapeutic strategy, have opened up new possibilities for the treatment of NDDs and brought new hope to patients.
Collapse
Affiliation(s)
- Ziying Liu
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Lin Cheng
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Wa Cao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Respiratory Medicine, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Chunxiao Shen
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Yuemin Qiu
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Chuan Li
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China
| | - Yinyi Xiong
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Rehabilitation, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China
| | - Seung Bum Yang
- Department of Medical Non-commissioned Officer, Wonkwang Health Science University Iksan-si, Jeollabuk-do 54538, South Korea
| | - Zhiying Chen
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Xiaoping Yin
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China; Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China.
| | - Xiaorong Zhang
- Department of Pathology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi 332000, China; Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi 332000, China.
| |
Collapse
|
41
|
Chan YT, Zhang C, Wu J, Lu P, Xu L, Yuan H, Feng Y, Chen ZS, Wang N. Biomarkers for diagnosis and therapeutic options in hepatocellular carcinoma. Mol Cancer 2024; 23:189. [PMID: 39242496 PMCID: PMC11378508 DOI: 10.1186/s12943-024-02101-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 09/09/2024] Open
Abstract
Liver cancer is a global health challenge, causing a significant social-economic burden. Hepatocellular carcinoma (HCC) is the predominant type of primary liver cancer, which is highly heterogeneous in terms of molecular and cellular signatures. Early-stage or small tumors are typically treated with surgery or ablation. Currently, chemotherapies and immunotherapies are the best treatments for unresectable tumors or advanced HCC. However, drug response and acquired resistance are not predictable with the existing systematic guidelines regarding mutation patterns and molecular biomarkers, resulting in sub-optimal treatment outcomes for many patients with atypical molecular profiles. With advanced technological platforms, valuable information such as tumor genetic alterations, epigenetic data, and tumor microenvironments can be obtained from liquid biopsy. The inter- and intra-tumoral heterogeneity of HCC are illustrated, and these collective data provide solid evidence in the decision-making process of treatment regimens. This article reviews the current understanding of HCC detection methods and aims to update the development of HCC surveillance using liquid biopsy. Recent critical findings on the molecular basis, epigenetic profiles, circulating tumor cells, circulating DNAs, and omics studies are elaborated for HCC diagnosis. Besides, biomarkers related to the choice of therapeutic options are discussed. Some notable recent clinical trials working on targeted therapies are also highlighted. Insights are provided to translate the knowledge into potential biomarkers for detection and diagnosis, prognosis, treatment response, and drug resistance indicators in clinical practice.
Collapse
Affiliation(s)
- Yau-Tuen Chan
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Cheng Zhang
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Junyu Wu
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Pengde Lu
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Lin Xu
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Hongchao Yuan
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Zhe-Sheng Chen
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
42
|
Lin J, Lyu Z, Feng H, Xie H, Peng J, Zhang W, Zheng J, Zheng J, Pan Z, Li Y. CircPDIA3/miR-449a/XBP1 feedback loop curbs pyroptosis by inhibiting palmitoylation of the GSDME-C domain to induce chemoresistance of colorectal cancer. Drug Resist Updat 2024; 76:101097. [PMID: 38861804 DOI: 10.1016/j.drup.2024.101097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 02/04/2024] [Accepted: 05/22/2024] [Indexed: 06/13/2024]
Abstract
Although oxaliplatin (OXA) is widely used in the frontline treatment of colorectal cancer (CRC), CRC recurrence is commonly observed due to OXA resistance. OXA resistance is associated with a number of factors, including abnormal regulation of pyroptosis. It is therefore important to elucidate the abnormal regulatory mechanism underlying pyroptosis. Here, we identified that the circular RNA circPDIA3 played an important role in chemoresistance in CRC. CircPDIA3 could induce chemoresistance in CRC by inhibiting pyroptosis both in vitro and in vivo. Mechanistically, RIP, RNA pull-down and co-IP assays revealed that circPDIA3 directly bonded to the GSDME-C domain, subsequently enhanced the autoinhibitory effect of the GSDME-C domain through blocking the GSDME-C domain palmitoylation by ZDHHC3 and ZDHHC17, thereby restraining pyroptosis. Additionally, it was found that the circPDIA3/miR-449a/XBP1 positive feedback loop increased the expression of circPDIA3 to induce chemoresistance. Furthermore, our clinical data and patient-derived tumor xenograft (PDX) models supported the positive association of circPDIA3 with development of chemoresistance in CRC patients. Taken together, our findings demonstrated that circPDIA3 could promote chemoresistance by amplifying the autoinhibitory effect of the GSDME-C domain through inhibition of the GSDME-C domain palmitoylation in CRC. This study provides novel insights into the mechanism of circRNA in regulating pyroptosis and providing a potential therapeutic target for reversing chemoresistance of CRC.
Collapse
Affiliation(s)
- Jiatong Lin
- School of Medicine South China University of Technology, Guangzhou 510006, China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zejian Lyu
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Huolun Feng
- School of Medicine South China University of Technology, Guangzhou 510006, China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Huajie Xie
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jingwen Peng
- Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, SunYat-sen University, Guangzhou 510120, China
| | - Weifu Zhang
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Guangdong Medical University, Dongguan 523808, China
| | - Jun Zheng
- Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou 510630, China; Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital of Sun Yat-sen University, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou 510630, China.
| | - Jiabin Zheng
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Zihao Pan
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| | - Yong Li
- School of Medicine South China University of Technology, Guangzhou 510006, China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| |
Collapse
|
43
|
Nedaeinia R, Najafgholian S, Salehi R, Goli M, Ranjbar M, Nickho H, Haghjooy Javanmard S, A Ferns G, Manian M. The role of cancer-associated fibroblasts and exosomal miRNAs-mediated intercellular communication in the tumor microenvironment and the biology of carcinogenesis: a systematic review. Cell Death Discov 2024; 10:380. [PMID: 39187523 PMCID: PMC11347635 DOI: 10.1038/s41420-024-02146-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024] Open
Abstract
CAFs (cancer-associated fibroblasts) are highly flexible cells of the cancer microenvironment. They produce the extracellular matrix (ECM) constituents that form the structure of the tumor stroma but are also a source of metabolites, growth factors, chemokines, and exosomes that impact every aspect of the tumor, including its response to treatment. It is believed that exosomal miRNAs facilitate intercellular signaling, which is essential for the development of cancer. The role of miRNAs and CAFs in the tumor microenvironment (TME) and carcinogenesis is reviewed in this paper. The preferred reporting items for systematic reviews and meta-analyses (PRISMA) 2020 guidelines were used to perform a systematic review. Several databases, including Web of Science, Medline, Embase, Cochrane Library, and Scopus, were searched using the following keywords: CAFs, CAF, cancer-associated fibroblasts, stromal fibroblasts, miRNA, exosomal miRNAs, exosome and similar terms. We identified studies investigating exosomal miRNAs and CAFs in the TME and their role in carcinogenesis. A total of 12,572 papers were identified. After removing duplicates (n = 3803), 8774 articles were screened by title and abstract. Of these, 421 were excluded from further analysis. It has been reported that if exosomal miRNAs in CAFs are not functioning correctly, this may influence the secretory phenotype of tip cells and contribute to increased tumor invasiveness, tumor spread, decreased treatment efficacy, and a poorer prognosis. Under their influence, normal fibroblasts (NFs) are transformed into CAFs. Furthermore, they participate in metabolic reprogramming, which allows for fast proliferation of the cancer cell population, adaptation to growing energy demands, and the capacity to avoid immune system identification.
Collapse
Affiliation(s)
- Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Simin Najafgholian
- Department of Emergency Medicine, School of Medicine, Valiasr Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Goli
- Department of Food Science and Technology, Laser and Biophotonics in Biotechnologies Research Center, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Maryam Ranjbar
- Advanced Materials Research Center, Department of Materials Engineering, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Hamid Nickho
- Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A Ferns
- Brighton and Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Mostafa Manian
- Department of Medical Laboratory Science, Faculty of Medical Science Kermanshah Branch, Islamic Azad University, Kermanshah, Iran.
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
44
|
Huang J, Cong F, Zhao Y, Chen J, Luo T, Tang W. The circular RNA circ_0001742 regulates colorectal carcinoma proliferation and migration via the MicroRNA-431-5p/ALG8 axis. Heliyon 2024; 10:e34660. [PMID: 39170557 PMCID: PMC11336282 DOI: 10.1016/j.heliyon.2024.e34660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Background Accumulating studies have found that circular RNAs (circRNAs) have a regulatory effect in a variety of tumors. However, to date, the relationship between specific circRNAs and colorectal cancer (CRC) remains elusive. Methods An RNA-sequencing method based on different metastatic potential of CRC cell lines was applied to evaluate the circRNA expression profile. Additionally, we conducted a series of experiments to assess the relationship between circRNAs and CRC progression. Results Circ_0001742 was upregulated in CRC cells with high metastatic potential, and circ_0001742 overexpression was observed to facilitate proliferation, migration and metastasis while knockdown will inhibit. More importantly, we found that circ_0001742 acted as a sponge for miR-431-5p, thus affecting ALG8 levels and the development of CRC. Conclusions This study demonstrated an essential function for the circ_0001742/miR-431-5p/ALG8 axis in CRC development, and it may be a promising therapeutic target for CRC.
Collapse
Affiliation(s)
- Jiahao Huang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Fengyun Cong
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| | - Yang Zhao
- Department of Radiology, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
| | - Jinglian Chen
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| | - Tao Luo
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| | - Weizhong Tang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital, Guangxi Medical University, Nanning, China
- Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| |
Collapse
|
45
|
Qian S, Zheng C, Wu Y, Huang H, Wu G, Zhang J. Targeted therapy for leukemia based on nanomaterials. Heliyon 2024; 10:e34951. [PMID: 39144922 PMCID: PMC11320317 DOI: 10.1016/j.heliyon.2024.e34951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/21/2024] [Accepted: 07/18/2024] [Indexed: 08/16/2024] Open
Abstract
Leukemia is a kind of hematopoietic stem cell malignant clonal disease. Drug therapy is the core treatment strategy for leukemia, but the current therapeutic drugs have defects such as low bioavailability, large adverse reactions and inconvenient intravenous administration. Targeted therapy can combine drugs with specific carcinogenic sites on cells to kill cancer cells and avoid damage to normal cells, which has gradually become the mainstream method of leukemia treatment. In addition, nanomedicine delivery systems can significantly improve drug efficacy through controlled size and targeted optimization of drug delivery by modification strategies. Therefore, the targeted treatment of leukemia based on nanomaterials has great research value and application prospect. This paper gives an overview of the current therapeutic strategies for leukemia, and then reviews the cutting-edge targeted therapeutic nanomaterials for leukemia, including organic nanomaterials (mainly carbon-based nanomaterials, lipid materials, polymers, etc.) and inorganic nanomaterials (mainly noble metal nanoparticles, magnetic nanoparticles, hollow mesoporous materials, etc.). The challenges and prospects for the future development of targeted nanomaterials in the treatment of leukemia are also briefly reviewed.
Collapse
Affiliation(s)
- Suying Qian
- Department of Hematology and Oncology, Ningbo No.2 Hospital, Ningbo, 315000, China
| | - Cuiping Zheng
- Department of Hematology and Oncology, Wenzhou Central Hospital, Wenzhou, 325099, China
| | - Yanfang Wu
- Department of Hematopathology, The First People's Hospital of Fuyang, Hangzhou, 311499, China
| | - Huiyan Huang
- Department of Hematopathology, The First People's Hospital of Fuyang, Hangzhou, 311499, China
| | - Gongqiang Wu
- Department of Hematology and Oncology, Dongyang People's Hospital, Jinhua, 322103, China
| | - Junyu Zhang
- Department of Hematopathology, Lishui Central Hospital, Lishui, 323020, China
| |
Collapse
|
46
|
Li S, Zhang H, Yu H, Wu Y, Yan L, Guan X, Dong B, Zhao M, Tian X, Hao C, Wu J. Combination of eribulin and anlotinib exerts synergistic cytotoxicity in retroperitoneal liposarcoma by inducing endoplasmic reticulum stress. Cell Death Discov 2024; 10:355. [PMID: 39117615 PMCID: PMC11310505 DOI: 10.1038/s41420-024-02103-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 07/11/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Primary retroperitoneal liposarcoma (RLPS) is a rare heterogeneous tumor occurring within retroperitoneal space, and its overall survival has not improved much in the past few decades. Based on a small-sample clinical practice at our center, patients with RLPS can greatly benefit from anlotinib and eribulin combination. In this study, we investigated the combinational effect of anlotinib and eribulin on RLPS. In vitro experiments revealed that a low dose of anlotinib significantly enhances the cytotoxic effects of eribulin, leading to a remarkable suppression of RLPS cell proliferation, viability, colony formation, migration, and cell-cycle progression compared to individual drug treatments. At the organoid level, the combination treatment causes the spheroids in Matrigel to disintegrate earlier than the single-drug group. In vivo, RLPS patient-derived xenograft (PDX) models demonstrated that the combination of these two drugs can obviously exert a safe and effective anti-tumor effect. Through transcriptome analysis, we uncovered and validated that the synergistic effect mainly is induced by the endoplasmic reticulum stress (ERS) pathway both in vitro and in vivo. Further analyses indicate that anlotinib plus eribulin treatment results in micro-vessel density and PD-L1 expression alterations, suggesting a potential impact on the tumor microenvironment. This study extensively explored the combination regimen at multiple levels and its underlying molecular mechanism in RLPS, thus providing a foundation for translational medicine research.
Collapse
Affiliation(s)
- Shuquan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Hongtao Zhang
- Guowen (Changchun) International Hospital, Changchun, Jilin Province, China
| | - Hao Yu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yifan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Liang Yan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaoya Guan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Bin Dong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, China
| | - Min Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiuyun Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jianhui Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
47
|
Fu M, Gao Q, Xiao M, Li RF, Sun XY, Li SL, Peng X, Ge XY. Extracellular Vesicles Containing circMYBL1 Induce CD44 in Adenoid Cystic Carcinoma Cells and Pulmonary Endothelial Cells to Promote Lung Metastasis. Cancer Res 2024; 84:2484-2500. [PMID: 38657100 DOI: 10.1158/0008-5472.can-23-3508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/13/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Adenoid cystic carcinoma (ACC) is a rare malignant epithelial neoplasm that arises in secretory glands and commonly metastasizes to the lungs. MYBL1 is frequently overexpressed in ACC and has been suggested to be a driver of the disease. In this study, we identified a circular RNA (circRNA) derived from MYBL1 pre-mRNA that was accompanied by the overexpression of MYBL1 in ACC. Overexpression of circMYBL1 was correlated with increased lung metastasis and poor overall survival in patients with ACC. Ectopic circMYBL1 overexpression promoted malignant phenotypes and lung metastasis of ACC cells. Mechanistically, circMYBL1 formed a circRNA-protein complex with CCAAT enhancer-binding protein β (CEBPB), which inhibited ubiquitin-mediated degradation and promoted nuclear translocation of CEBPB. In the nucleus, circMYBL1 increased the binding of CEBPB to the CD44 promoter region and enhanced its transcription. In addition, circMYBL1 was enriched in small extracellular vesicles (sEV) isolated from the plasma of patients with ACC. Treatment with sEVs containing circMYBL1 in sEVs enhanced prometastatic phenotypes of ACC cells, elevated the expression of CD44 in human pulmonary microvascular endothelial cells (HPMEC), and enhanced the adhesion between HPMECs and ACC cells. Moreover, circMYBL1 encapsulated in sEVs increased the arrest of circulating ACC cells in the lung and enhanced lung metastatic burden. These data suggest that circMYBL1 is a tumor-promoting circRNA that could serve as a potential biomarker and therapeutic target for ACC. Significance: circMYBL1 stabilizes CEBPB and upregulates CD44 to promote adhesion between cancer cells and endothelial cells and enables lung metastasis of adenoid cystic carcinoma, suggesting that inhibition of this axis could improve patient outcomes.
Collapse
MESH Headings
- Humans
- Lung Neoplasms/secondary
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Hyaluronan Receptors/metabolism
- Hyaluronan Receptors/genetics
- Carcinoma, Adenoid Cystic/pathology
- Carcinoma, Adenoid Cystic/metabolism
- Carcinoma, Adenoid Cystic/genetics
- Carcinoma, Adenoid Cystic/secondary
- Mice
- Animals
- Extracellular Vesicles/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- RNA, Circular/genetics
- RNA, Circular/metabolism
- CCAAT-Enhancer-Binding Protein-beta/metabolism
- CCAAT-Enhancer-Binding Protein-beta/genetics
- Cell Line, Tumor
- Female
- Mice, Nude
- Male
- Gene Expression Regulation, Neoplastic
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins/genetics
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Min Fu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, PR China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, Beijing, PR China
- National Clinical Research Center for Oral Diseases, Beijing, PR China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, PR China
| | - Qian Gao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Mian Xiao
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Rui-Feng Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xin-Yi Sun
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Sheng-Lin Li
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xin Peng
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, PR China
| | - Xi-Yuan Ge
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, PR China
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, PR China
- National Center for Stomatology, Beijing, PR China
- National Clinical Research Center for Oral Diseases, Beijing, PR China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, PR China
| |
Collapse
|
48
|
Liu C, Zou Z, Lu S, Jin K, Shen Y, Huang T, Li W, Zhou G. CircPKN2 promotes ferroptosis in bladder cancer by promoting the ubiquitination of Stearoyl-CoA Desaturase 1. Cancer Gene Ther 2024; 31:1251-1265. [PMID: 38802550 DOI: 10.1038/s41417-024-00784-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 05/05/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
Bladder cancer (BC) is one of the most common malignancies in the male urinary system and currently lacks an optimal treatment strategy. To elucidate the pathogenic mechanisms of BC from the perspective of circular RNAs, we conducted this study. Building upon our previous research, a novel circRNA, circPKN2, captured our interest due to its significant downregulation in BC, and its close association with the prognosis of BC patients. Our research findings indicate that circPKN2 can inhibit the proliferation and migration of BC cells in vitro. Furthermore, we discovered that circPKN2 exerts its anti-cancer effects in BC by promoting ferroptosis. Mechanistic studies revealed that circPKN2 recruits STUB1 to facilitate the ubiquitination of SCD1, thereby suppressing the WNT pathway and promoting ferroptosis in BC. Additionally, our research unveiled the regulatory role of the splicing factor QKI in the biogenesis of circPKN2. Animal studies demonstrated that circPKN2 enhances ferroptosis in BC cells in vivo, inhibiting tumor growth and metastasis. The discovery of the anti-cancer factor circPKN2 holds promise for providing new therapeutic targets in the prevention and treatment of BC.
Collapse
Affiliation(s)
- Changkun Liu
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
| | - Zhuo Zou
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Graduate School, Dalian Medical University, Dalian, China
| | - Shengming Lu
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Kun Jin
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Ye Shen
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Tianbao Huang
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Weijian Li
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Guangchen Zhou
- Department of Urology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
| |
Collapse
|
49
|
Li Q, Zhang Y, Jin P, Chen Y, Zhang C, Geng X, Mun KS, Phang KC. New insights into the potential of exosomal circular RNAs in mediating cancer chemotherapy resistance and their clinical applications. Biomed Pharmacother 2024; 177:117027. [PMID: 38925018 DOI: 10.1016/j.biopha.2024.117027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/28/2024] Open
Abstract
Chemotherapy resistance typically leads to tumour recurrence and is a major obstacle to cancer treatment. Increasing numbers of circular RNAs (circRNAs) have been confirmed to be abnormally expressed in various tumours, where they participate in the malignant progression of tumours, and play important roles in regulating the sensitivity of tumours to chemotherapy drugs. As exosomes mediate intercellular communication, they are rich in circRNAs and exhibit a specific RNA cargo sorting mechanism. By carrying and delivering circRNAs, exosomes can promote the efflux of chemotherapeutic drugs and reduce intracellular drug concentrations in recipient cells, thus affecting the cell cycle, apoptosis, autophagy, angiogenesis, invasion and migration. The mechanisms that affect the phenotype of tumour stem cells, epithelial-mesenchymal transformation and DNA damage repair also mediate chemotherapy resistance in many tumours. Exosomal circRNAs are diagnostic biomarkers and potential therapeutic targets for reversing chemotherapy resistance in tumours. Currently, the rise of new fields, such as machine learning and artificial intelligence, and new technologies such as biosensors, multimolecular diagnostic systems and platforms based on circRNAs, as well as the application of exosome-based vaccines, has provided novel ideas for precision cancer treatment. In this review, the recent progress in understanding how exosomal circRNAs mediate tumour chemotherapy resistance is reviewed, and the potential of exosomal circRNAs in tumour diagnosis, treatment and immune regulation is discussed, providing new ideas for inhibiting tumour chemotherapy resistance.
Collapse
Affiliation(s)
- Qiang Li
- School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China; Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Yuhao Zhang
- Department of Neurosurgery, Zhejiang Provincial People's Hospital, Affiliated to Hangzhou Medical College, Hangzhou, Zhejiang 310000, China
| | - Peikan Jin
- School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Yepeng Chen
- School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Chuchu Zhang
- School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China
| | - Xiuchao Geng
- School of Medicine, Taizhou University, Taizhou, Zhejiang 318000, China.
| | - Kein Seong Mun
- Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Kean Chang Phang
- Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| |
Collapse
|
50
|
Wang D, Chen D, Liang L, Hu J. The circZEB1/miR-337-3p/ OGT axis mediates angiogenesis and metastasis via O-GlcNAcylation and up-regulating YBX1 in breast cancer. Heliyon 2024; 10:e34079. [PMID: 39114035 PMCID: PMC11305230 DOI: 10.1016/j.heliyon.2024.e34079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 07/03/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND A growing corpus of research has revealed that circular RNAs (circRNAs) have become increasingly important for the growth of malignancies in recent years. CircRNAs as ideal candidates for breast cancer (BC) therapeutic targets is still absent. METHODS In our study, the dysregulated circRNAs in BC progression were explored, we analysed the BC's circRNA expression profiles using publicly available datasets (GSE101124 and GSE101122). The expression of circZEB1 in BC and cell lines was investigated by qPCR. RNase and actinomycin D were used to examine the features of circZEB1. The function of circZEB1 was subsequently investigated through the utilisation of colony formation, tube formation, transwell assays, and xenograft animal models.RNA immunoprecipitation (RIP), luciferase reporter assays, immunoprecipitation (co-IP) test in conjunction with LC-MS, and ChIP-seq assay to investigate the molecular mechanism underlying the biological activity of circZEB1 in BC. RESULTS Among the circRNAs, we were particularly interested in hsa_circ_0000228, which is spliced from the oncogene ZEB1. In BC cell lines, CircZEB1 expression was upregulated. CircZEB1 knockdown prevented BC cells from migrating and invading, as well as HUVECs from forming tubes and developing. By sponging miR-337-3p, functional testing revealed that circZEB1 promoted O-GlcNAcylation, increased YBX1, and OGT expression. Moreover, circZEB1 overexpression is reversible, in contrast to YBX1 knockdown, which mostly results in the downregulation of multiple oncogenes. CONCLUSION Our study indicate that circZEB1 had oncogenic function in BC by focusing on circZEB1/miR-337-3p/OGT and YBX1. It might be inferred that circZEB1 could be a promising new target for BC treatment.
Collapse
Affiliation(s)
- Dongying Wang
- Molecular Genetics Laboratory, Yiwu Maternity and Children Hospital, Jinhua, 321000, China
| | - Dengyi Chen
- Department of Clinical Laboratory, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, 441000, China
| | - Leilei Liang
- Zhejiang Cancer Hospital, Hangzhou, 310011, China
| | - Jialei Hu
- Molecular Genetics Laboratory, Yiwu Maternity and Children Hospital, Jinhua, 321000, China
| |
Collapse
|